101
|
Ma WY, Wang SS, Wu QL, Zhou X, Chu SF, Chen NH. The versatile role of TREM2 in regulating of microglia fate in the ischemic stroke. Int Immunopharmacol 2022; 109:108733. [PMID: 35525233 DOI: 10.1016/j.intimp.2022.108733] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022]
Abstract
Microglia are the earliest activated and the longest lasting immune cells after stroke, and they participate in almost all the pathological reactions after stroke. However, their regulatory mechanism has not been fully elucidated. Triggering receptor expressed on myeloid cells-2 (TREM2) is a cell surface receptor that is mainly expressed in microglia of the central nervous system. The receptor plays an important role in regulating microglia energy metabolism and phenotypic transformation. At present, TREM2 has been developed as a potential target for AD, coronary atherosclerosis and other diseases. However, TREM2 does not provide a systematic summary of the functional transformation and intrinsic molecular mechanisms of microglia after stroke. In this paper, we have summarized the functional changes of TREM2 in microglia after stroke in recent years, and found that TREM2 has important effects on energy metabolism, phagocytosis and anti-inflammatory function of microglia after stroke, suggesting that TREM2 is a potential therapeutic target for the treatment of stroke.
Collapse
Affiliation(s)
- Wen-Yu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Sha-Sha Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qing-Lin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xin Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medical & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
102
|
Neuner SM, Telpoukhovskaia M, Menon V, O'Connell KMS, Hohman TJ, Kaczorowski CC. Translational approaches to understanding resilience to Alzheimer's disease. Trends Neurosci 2022; 45:369-383. [PMID: 35307206 PMCID: PMC9035083 DOI: 10.1016/j.tins.2022.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/07/2022] [Accepted: 02/23/2022] [Indexed: 10/18/2022]
Abstract
Individuals who maintain cognitive function despite high levels of Alzheimer's disease (AD)-associated pathology are said to be 'resilient' to AD. Identifying mechanisms underlying resilience represents an exciting therapeutic opportunity. Human studies have identified a number of molecular and genetic factors associated with resilience, but the complexity of these cohorts prohibits a complete understanding of which factors are causal or simply correlated with resilience. Genetically and phenotypically diverse mouse models of AD provide new and translationally relevant opportunities to identify and prioritize new resilience mechanisms for further cross-species investigation. This review will discuss insights into resilience gained from both human and animal studies and highlight future approaches that may help translate these insights into therapeutics designed to prevent or delay AD-related dementia.
Collapse
Affiliation(s)
- Sarah M Neuner
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kristen M S O'Connell
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Tufts University, School of Medicine, Graduate School of Biomedical Sciences, Boston, MA 02111, USA; The University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME 04469, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Catherine C Kaczorowski
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Tufts University, School of Medicine, Graduate School of Biomedical Sciences, Boston, MA 02111, USA; The University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME 04469, USA.
| |
Collapse
|
103
|
Leng F, Zhan Z, Sun Y, Liu F, Edison P, Sun Y, Wang Z. Cerebrospinal Fluid sTREM2 Has Paradoxical Association with Brain Structural Damage Rate in Early- and Late-Stage Alzheimer’s Disease. J Alzheimers Dis 2022; 88:117-126. [PMID: 35491791 DOI: 10.3233/jad-220102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Recently it has been proposed that microglial response has a stage-dependent effect on the progression of Alzheimer’s disease (AD). Cerebrospinal fluid (CSF) sTREM2 has emerged as a promising microglial activation marker. Objective: To test the stage-dependent role of microglia by studying the association between baseline sTREM2 and dynamic brain structural changes in AD and mild cognitive impairment (MCI) patients. Methods: 22 amyloid-β-positive (A+) and tau-positive (T+) AD and 24 A+T+MCI patients were identified from the Alzheimer’s Disease Neuroimaging Initiative. The patients had baseline CSF amyloid-β, phosphorylated-tau, and sTREM2, and were followed up for at least one year by T1-weighted and diffusion tensor imaging scans. Gray matter volumes and white matter microstructural integrity were evaluated. Linear mixed models were applied to analyze how baseline sTREM2 may influence the rate of brain structural changes while adjusting for the effects of age, APOE4 status, and the CSF core markers. Results: In A+T+AD patients, baseline CSF sTREM2 was associated with faster mean diffusivity increase in the bilateral posterior corona radiata and right superior longitudinal fasciculus. In A+T+MCI patients, baseline CSF sTREM2 was associated slower gray matter volumetric loss in parahippocampal gyrus, left fusiform cortex, left middle temporal gyrus, and left lateral occipital cortex. Baseline CSF sTREM2 also had a protective effect against mean diffusivity increase in right inferior fronto-occipital fasciculus, left superior longitudinal fasciculus, left forceps minor, and left uncinate fasciculus. Conclusion: Microglial activation at early stage might have a protective effect against neurodegeneration, while at late stage it might facilitate AD. Future efforts on modulating microglial activation could be promising, given a carefully selected time window for intervention.
Collapse
Affiliation(s)
- Fangda Leng
- Department of Neurology, Peking University First Hospital, Peking University, Beijing, China
| | - Zhenying Zhan
- Department of Neurology, Pujiang Branch, The First Affiliated Hospital of Zhejiang University, Zhejiang, China
| | - Yunchuang Sun
- Department of Neurology, Peking University First Hospital, Peking University, Beijing, China
| | - Fang Liu
- Department of Neurology, Tsinghua University First Hospital, Tsinghua University, Beijing, China
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, UK
| | - Yongan Sun
- Department of Neurology, Peking University First Hospital, Peking University, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Peking University, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | | |
Collapse
|
104
|
Quiroga IY, Cruikshank AE, Bond ML, Reed KSM, Evangelista BA, Tseng JH, Ragusa JV, Meeker RB, Won H, Cohen S, Cohen TJ, Phanstiel DH. Synthetic amyloid beta does not induce a robust transcriptional response in innate immune cell culture systems. J Neuroinflammation 2022; 19:99. [PMID: 35459147 PMCID: PMC9034485 DOI: 10.1186/s12974-022-02459-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease that impacts nearly 400 million people worldwide. The accumulation of amyloid beta (Aβ) in the brain has historically been associated with AD, and recent evidence suggests that neuroinflammation plays a central role in its origin and progression. These observations have given rise to the theory that Aβ is the primary trigger of AD, and induces proinflammatory activation of immune brain cells (i.e., microglia), which culminates in neuronal damage and cognitive decline. To test this hypothesis, many in vitro systems have been established to study Aβ-mediated activation of innate immune cells. Nevertheless, the transcriptional resemblance of these models to the microglia in the AD brain has never been comprehensively studied on a genome-wide scale. METHODS We used bulk RNA-seq to assess the transcriptional differences between in vitro cell types used to model neuroinflammation in AD, including several established, primary and iPSC-derived immune cell lines (macrophages, microglia and astrocytes) and their similarities to primary cells in the AD brain. We then analyzed the transcriptional response of these innate immune cells to synthetic Aβ or LPS and INFγ. RESULTS We found that human induced pluripotent stem cell (hIPSC)-derived microglia (IMGL) are the in vitro cell model that best resembles primary microglia. Surprisingly, synthetic Aβ does not trigger a robust transcriptional response in any of the cellular models analyzed, despite testing a wide variety of Aβ formulations, concentrations, and treatment conditions. Finally, we found that bacterial LPS and INFγ activate microglia and induce transcriptional changes that resemble many, but not all, aspects of the transcriptomic profiles of disease associated microglia (DAM) present in the AD brain. CONCLUSIONS These results suggest that synthetic Aβ treatment of innate immune cell cultures does not recapitulate transcriptional profiles observed in microglia from AD brains. In contrast, treating IMGL with LPS and INFγ induces transcriptional changes similar to those observed in microglia detected in AD brains.
Collapse
Affiliation(s)
- I Y Quiroga
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - A E Cruikshank
- Postbaccalaureate Research Education Program, University of North Carolina, Chapel Hill, NC, USA
| | - M L Bond
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, USA
| | - K S M Reed
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, USA
| | - B A Evangelista
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - J H Tseng
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - J V Ragusa
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - R B Meeker
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - H Won
- Department of Genetics and Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - S Cohen
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - T J Cohen
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - D H Phanstiel
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA.
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, USA.
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
105
|
Mankhong S, Kim S, Lee S, Kwak HB, Park DH, Joa KL, Kang JH. Development of Alzheimer’s Disease Biomarkers: From CSF- to Blood-Based Biomarkers. Biomedicines 2022; 10:biomedicines10040850. [PMID: 35453600 PMCID: PMC9025524 DOI: 10.3390/biomedicines10040850] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
In the 115 years since the discovery of Alzheimer’s disease (AD), our knowledge, diagnosis, and therapeutics have significantly improved. Biomarkers are the primary tools for clinical research, diagnostics, and therapeutic monitoring in clinical trials. They provide much insightful information, and while they are not clinically used routinely, they help us to understand the mechanisms of this disease. This review charts the journey of AD biomarker discovery and development from cerebrospinal fluid (CSF) amyloid-beta 1-42 (Aβ42), total tau (T-tau), and phosphorylated tau (p-tau) biomarkers and imaging technologies to the next generation of biomarkers. We also discuss advanced high-sensitivity assay platforms for CSF Aβ42, T-tau, p-tau, and blood analysis. The recently proposed Aβ deposition/tau biomarker/neurodegeneration or neuronal injury (ATN) scheme might facilitate the definition of the biological status underpinning AD and offer a common language among researchers across biochemical biomarkers and imaging. Moreover, we highlight blood-based biomarkers for AD that offer a scalable alternative to CSF biomarkers through cost-saving and reduced invasiveness, and may provide an understanding of disease initiation and development. We discuss different groups of blood-based biomarker candidates, their advantages and limitations, and paths forward, from identification and analysis to clinical validation. The development of valid blood-based biomarkers may facilitate the implementation of future AD therapeutics and diagnostics.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.)
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
| | - Sujin Kim
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.)
| | - Seongju Lee
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Department of Anatomy, College of Medicine, Inha University, Incheon 22212, Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Kyung-Lim Joa
- Department of Physical & Rehabilitation Medicine, College of Medicine, Inha University, Incheon 22212, Korea;
| | - Ju-Hee Kang
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.)
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea; (S.L.); (H.-B.K.); (D.-H.P.)
- Correspondence: ; Tel.: +82-32-860-9872
| |
Collapse
|
106
|
Dong MH, Zhou LQ, Tang Y, Chen M, Xiao J, Shang K, Deng G, Qin C, Tian DS. CSF sTREM2 in neurological diseases: a two-sample Mendelian randomization study. J Neuroinflammation 2022; 19:79. [PMID: 35382840 PMCID: PMC8985278 DOI: 10.1186/s12974-022-02443-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background Soluble triggering receptor expressed on myeloid cells 2 (sTREM2) in cerebrospinal fluid (CSF) has been described as a biomarker for microglial activation, which were observed increased in a variety of neurological disorders. Objective Our objective was to explore whether genetically determined CSF sTREM2 levels are causally associated with different neurological diseases by conducting a two-sample Mendelian randomization (MR) study. Methods Single nucleotide polymorphisms significantly associated with CSF sTREM2 levels were selected as instrumental variables to estimate the causal effects on clinically common neurological diseases, including stroke, Alzheimer’s diseases, Parkinson’s diseases, amyotrophic lateral sclerosis, multiple sclerosis, and epilepsy and their subtypes. Summary-level statistics of both exposure and outcomes were applied in an MR framework. Results Genetically predicted per 1 pg/dL increase of CSF sTREM2 levels was associated with higher risk of multiple sclerosis (OR = 1.038, 95%CI = 1.014–1.064, p = 0.002). Null association was found in risk of other included neurological disorders. Conclusions These findings provide support for a potential causal relationship between elevated CSF sTREM2 levels and higher risk of multiple sclerosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02443-9.
Collapse
Affiliation(s)
- Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
107
|
sTREM2 mediates the associations of minimal depressive symptoms with amyloid pathology in prodromal Alzheimer's disease: The CABLE study. Transl Psychiatry 2022; 12:140. [PMID: 35379792 PMCID: PMC8980028 DOI: 10.1038/s41398-022-01910-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/15/2022] Open
Abstract
The effects of microglial activation on the associations between depression and Alzheimer's disease (AD) are still unclear. TREM2 gene plays a pivotal role in microglial activation, has been identified as a risk factor for AD. In this work, we aimed to assess the interrelationships of soluble TREM2 (sTREM2) level in cerebrospinal fluid (CSF), minimal depressive symptoms (MDSs), and CSF amyloid markers. The linear regression analyses were conducted on 796 cognitively unimpaired participants from the CABLE (Chinese Alzheimer's Biomarker and LifestylE) study. Causal mediation analyses with 10,000 bootstrapped iterations were used to test the mediation effects. In addition, similar statistical analyses were performed in subgroups stratified by sex, age, and APOE ε4 carrier status. In total subjects, MDSs were associated with lower CSF sTREM2 levels (p < 0.0001), lower CSF amyloid markers (p < 0.0001), and poorer cognitive performance (MMSE, p = 0.0014). The influence of MDSs on CSF amyloid markers was partially mediated by CSF sTREM2 (proportion from 2.91 to 32.58%, p < 0.0001). And we found that the sTREM2-amyloid pathway partially mediated the effects of MDSs on cognition. Of note, exploratory subgroup analyses showed that the above influences of CSF sTREM2 were pronounced in the APOE ε4 (-) group. These results suggest that early depression is associated with amyloid pathology, which might be partly mediated by microglial activation, especially in the absence of APOE ε4.
Collapse
|
108
|
Morenas-Rodríguez E, Li Y, Nuscher B, Franzmeier N, Xiong C, Suárez-Calvet M, Fagan AM, Schultz S, Gordon BA, Benzinger TLS, Hassenstab J, McDade E, Feederle R, Karch CM, Schlepckow K, Morris JC, Kleinberger G, Nellgard B, Vöglein J, Blennow K, Zetterberg H, Ewers M, Jucker M, Levin J, Bateman RJ, Haass C. Soluble TREM2 in CSF and its association with other biomarkers and cognition in autosomal-dominant Alzheimer's disease: a longitudinal observational study. Lancet Neurol 2022; 21:329-341. [PMID: 35305339 PMCID: PMC8926925 DOI: 10.1016/s1474-4422(22)00027-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Therapeutic modulation of TREM2-dependent microglial function might provide an additional strategy to slow the progression of Alzheimer's disease. Although studies in animal models suggest that TREM2 is protective against Alzheimer's pathology, its effect on tau pathology and its potential beneficial role in people with Alzheimer's disease is still unclear. Our aim was to study associations between the dynamics of soluble TREM2, as a biomarker of TREM2 signalling, and amyloid β (Aβ) deposition, tau-related pathology, neuroimaging markers, and cognitive decline, during the progression of autosomal dominant Alzheimer's disease. METHODS We did a longitudinal analysis of data from the Dominantly Inherited Alzheimer Network (DIAN) observational study, which includes families with a history of autosomal dominant Alzheimer's disease. Participants aged over 18 years who were enrolled in DIAN between Jan 1, 2009, and July 31, 2019, were categorised as either carriers of pathogenic variants in PSEN1, PSEN2, and APP genes (n=155) or non-carriers (n=93). We measured amounts of cleaved soluble TREM2 using a novel immunoassay in CSF samples obtained every 2 years from participants who were asymptomatic (Clinical Dementia Rating [CDR]=0) and annually for those who were symptomatic (CDR>0). CSF concentrations of Aβ40, Aβ42, total tau (t-tau), and tau phosphorylated on threonine 181 (p-tau) were measured by validated immunoassays. Predefined neuroimaging measurements were total cortical uptake of Pittsburgh compound B PET (PiB-PET), cortical thickness in the precuneus ascertained by MRI, and hippocampal volume determined by MRI. Cognition was measured using a validated cognitive composite (including DIAN word list test, logical memory delayed recall, digit symbol coding test [total score], and minimental status examination). We based our statistical analysis on univariate and bivariate linear mixed effects models. FINDINGS In carriers of pathogenic variants, a high amyloid burden at baseline, represented by low CSF Aβ42 (β=-4·28 × 10-2 [SE 0·013], p=0·0012), but not high cortical uptake in PiB-PET (β=-5·51 × 10-3 [0·011], p=0·63), was the only predictor of an augmented annual rate of subsequent increase in soluble TREM2. Augmented annual rates of increase in soluble TREM2 were associated with a diminished rate of decrease in amyloid deposition, as measured by Aβ42 in CSF (r=0·56 [0·22], p=0·011), in presymptomatic carriers of pathogenic variants, and with diminished annual rate of increase in PiB-PET (r=-0·67 [0·25], p=0·0060) in symptomatic carriers of pathogenic variants. Presymptomatic carriers of pathogenic variants with annual rates of increase in soluble TREM2 lower than the median showed a correlation between enhanced annual rates of increase in p-tau in CSF and augmented annual rates of increase in PiB-PET signal (r=0·45 [0·21], p=0·035), that was not observed in those with rates of increase in soluble TREM2 higher than the median. Furthermore, presymptomatic carriers of pathogenic variants with rates of increase in soluble TREM2 above or below the median had opposite associations between Aβ42 in CSF and PiB-PET uptake when assessed longitudinally. Augmented annual rates of increase in soluble TREM2 in presymptomatic carriers of pathogenic variants correlated with decreased cortical shrinkage in the precuneus (r=0·46 [0·22]), p=0·040) and diminished cognitive decline (r=0·67 [0·22], p=0·0020). INTERPRETATION Our findings in autosomal dominant Alzheimer's disease position the TREM2 response within the amyloid cascade immediately after the first pathological changes in Aβ aggregation and further support the role of TREM2 on Aβ plaque deposition and compaction. Furthermore, these findings underpin a beneficial effect of TREM2 on Aβ deposition, Aβ-dependent tau pathology, cortical shrinkage, and cognitive decline. Soluble TREM2 could, therefore, be a key marker for clinical trial design and interpretation. Efforts to develop TREM2-boosting therapies are ongoing. FUNDING German Research Foundation, US National Institutes of Health.
Collapse
Affiliation(s)
- Estrella Morenas-Rodríguez
- German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany.
| | - Yan Li
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, USA
| | - Brigitte Nuscher
- German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St Louis, MO, USA
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Servei de Neurologia, Hospital del Mar Medical Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Madrid, Spain
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Stephanie Schultz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Eric McDade
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Regina Feederle
- German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Center, Munich, Germany; German Research Center for Environmental Health, Neuherberg, Germany
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Gernot Kleinberger
- German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Bengt Nellgard
- Department of Anesthesiology and Intensive Care, Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonathan Vöglein
- German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, University Hospital of Munich, Ludwig-Maximilians University, Munich, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Queens Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Hong Kong Special Administrative Region, China
| | - Michael Ewers
- German Center for Neurodegenerative Diseases, Munich, Germany; Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases, Tübingen, Germany; Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases, Munich, Germany; Department of Neurology, University Hospital of Munich, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
109
|
Smirnov D, Galasko D. Dynamics of neuroinflammation in Alzheimer's disease. Lancet Neurol 2022; 21:297-298. [PMID: 35305329 DOI: 10.1016/s1474-4422(22)00087-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Denis Smirnov
- Department of Neurosciences, UC San Diego, La Jolla, CA 92093-0948, USA
| | - Douglas Galasko
- Department of Neurosciences, UC San Diego, La Jolla, CA 92093-0948, USA.
| |
Collapse
|
110
|
Song C, Shi J, Zhang P, Zhang Y, Xu J, Zhao L, Zhang R, Wang H, Chen H. Immunotherapy for Alzheimer's disease: targeting β-amyloid and beyond. Transl Neurodegener 2022; 11:18. [PMID: 35300725 PMCID: PMC8932191 DOI: 10.1186/s40035-022-00292-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/25/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the elderly worldwide. However, the complexity of AD pathogenesis leads to discrepancies in the understanding of this disease, and may be the main reason for the failure of AD drug development. Fortunately, many ongoing preclinical and clinical studies will continually open up avenues to unravel disease mechanisms and guide strategies for AD diagnosis and drug development. For example, immunotherapeutic strategies targeting amyloid-β (Aβ) and tau proteins were once deemed almost certainly effective in clinical treatment due to the excellent preclinical results. However, the repeated failures of clinical trials on vaccines and humanized anti-Aβ and anti-tau monoclonal antibodies have resulted in doubts on this strategy. Recently, a new anti-Aβ monoclonal antibody (Aducanumab) has been approved by the US Food and Drug Administration, which brings us back to the realization that immunotherapy strategies targeting Aβ may be still promising. Meanwhile, immunotherapies based on other targets such as tau, microglia and gut-brain axis are also under development. Further research is still needed to clarify the forms and epitopes of targeted proteins to improve the accuracy and effectiveness of immunotherapeutic drugs. In this review, we focus on the immunotherapies based on Aβ, tau and microglia and their mechanisms of action in AD. In addition, we present up-to-date advances and future perspectives on immunotherapeutic strategies for AD.
Collapse
Affiliation(s)
- Chenghuan Song
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiyun Shi
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Pingao Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yongfang Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lanxue Zhao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Clinical Pharmacy, Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
111
|
Weidung B, Hemmingsson E, Olsson J, Sundström T, Blennow K, Zetterberg H, Ingelsson M, Elgh F, Lövheim H. VALZ-Pilot: High-dose valacyclovir treatment in patients with early-stage Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12264. [PMID: 35310522 PMCID: PMC8919248 DOI: 10.1002/trc2.12264] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/16/2021] [Accepted: 01/11/2022] [Indexed: 11/09/2022]
Abstract
Introduction Herpes simplex virus (HSV) may be involved in Alzheimer's disease (AD) pathophysiology. The antiviral valacyclovir inhibits HSV replication. Methods This phase-II pilot trial involved valacyclovir administration (thrice daily, 500 mg week 1, 1000 mg weeks 2-4) to persons aged ≥ 65 years with early-stage AD, anti-HSV immunoglobulin G, and apolipoprotein E ε4. Intervention safety, tolerability, feasibility, and effects on Mini-Mental State Examination (MMSE) scores and cerebrospinal fluid (CSF) biomarkers were evaluated. Results Thirty-two of 33 subjects completed the trial on full dosage. Eighteen percent experienced likely intervention-related mild, temporary adverse events. CSF acyclovir concentrations were mean 5.29 ± 2.31 μmol/L. CSF total tau and neurofilament light concentrations were unchanged; MMSE score and CSF soluble triggering receptor expressed on myeloid cells 2 concentrations increased (P = .02 and .03). Discussion Four weeks of high-dose valacyclovir treatment was safe, tolerable, and feasible in early-stage AD. Our findings may guide future trial design.
Collapse
Affiliation(s)
- Bodil Weidung
- Section of GeriatricsDepartment of Public Health and Caring SciencesUppsala UniversityUppsalaSweden
| | - Eva‐Stina Hemmingsson
- Department of Community Medicine and RehabilitationGeriatric MedicineUmeå UniversityUmeåSweden
| | - Jan Olsson
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
| | - Torbjörn Sundström
- Diagnostic RadiologyDepartment of Radiation SciencesUmeå UniversityUmeåSweden
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Martin Ingelsson
- Section of GeriatricsDepartment of Public Health and Caring SciencesUppsala UniversityUppsalaSweden
- Krembil Brain InstituteUniversity Health NetworkTorontoCanada
- Department of Medicine and Tanz Centre for Research in Neurodegenerative DiseasesUniversity of TorontoTorontoCanada
| | - Fredrik Elgh
- Department of Clinical MicrobiologyUmeå UniversityUmeåSweden
| | - Hugo Lövheim
- Department of Community Medicine and RehabilitationGeriatric MedicineUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicinUmeåSweden
| |
Collapse
|
112
|
Chen HH, Eteleeb A, Wang C, Fernandez MV, Budde JP, Bergmann K, Norton J, Wang F, Ebl C, Morris JC, Perrin RJ, Bateman RJ, McDade E, Xiong C, Goate A, Farlow M, Chhatwal J, Schofield PR, Chui H, Harari O, Cruchaga C, Ibanez L. Circular RNA detection identifies circPSEN1 alterations in brain specific to autosomal dominant Alzheimer's disease. Acta Neuropathol Commun 2022; 10:29. [PMID: 35246267 PMCID: PMC8895634 DOI: 10.1186/s40478-022-01328-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/07/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Autosomal-dominant Alzheimer's disease (ADAD) is caused by pathogenic mutations in APP, PSEN1, and PSEN2, which usually lead to an early age at onset (< 65). Circular RNAs are a family of non-coding RNAs highly expressed in the nervous system and especially in synapses. We aimed to investigate differences in brain gene expression of linear and circular transcripts from the three ADAD genes in controls, sporadic AD, and ADAD. METHODS We obtained and sequenced RNA from brain cortex using standard protocols. Linear counts were obtained using the TOPMed pipeline; circular counts, using python package DCC. After stringent quality control (QC), we obtained the counts for PSEN1, PSEN2 and APP genes. Only circPSEN1 passed QC. We used DESeq2 to compare the counts across groups, correcting for biological and technical variables. Finally, we performed in-silico functional analyses using the Circular RNA interactome website and DIANA mirPath software. RESULTS Our results show significant differences in gene counts of circPSEN1 in ADAD individuals, when compared to sporadic AD and controls (ADAD = 21, AD = 253, Controls = 23-ADADvsCO: log2FC = 0.794, p = 1.63 × 10-04, ADADvsAD: log2FC = 0.602, p = 8.22 × 10-04). The high gene counts are contributed by two circPSEN1 species (hsa_circ_0008521 and hsa_circ_0003848). No significant differences were observed in linear PSEN1 gene expression between cases and controls, indicating that this finding is specific to the circular forms. In addition, the high circPSEN1 levels do not seem to be specific to PSEN1 mutation carriers; the counts are also elevated in APP and PSEN2 mutation carriers. In-silico functional analyses suggest that circPSEN1 is involved in several pathways such as axon guidance (p = 3.39 × 10-07), hippo signaling pathway (p = 7.38 × 10-07), lysine degradation (p = 2.48 × 10-05) or Wnt signaling pathway (p = 5.58 × 10-04) among other KEGG pathways. Additionally, circPSEN1 counts were able to discriminate ADAD from sporadic AD and controls with an AUC above 0.70. CONCLUSIONS Our findings show the differential expression of circPSEN1 is increased in ADAD. Given the biological function previously ascribed to circular RNAs and the results of our in-silico analyses, we hypothesize that this finding might be related to neuroinflammatory events that lead or that are caused by the accumulation of amyloid-beta.
Collapse
Affiliation(s)
- Hsiang-Han Chen
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Abdallah Eteleeb
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Ciyang Wang
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Maria Victoria Fernandez
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - John P. Budde
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Kristy Bergmann
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Joanne Norton
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Fengxian Wang
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Curtis Ebl
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - John C. Morris
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Richard J. Perrin
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Randall J. Bateman
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Eric McDade
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Martin Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA USA
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Helena Chui
- Department of Neurology, Keck School of Medicine of University of Southern California, Los Angeles, CA USA
| | - Oscar Harari
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Genetics, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| | - Dominantly Inherited Alzheimer Network
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, 4444 Forest Park, Campus Box 8134, Saint Louis, MO 63110 USA
- NeuroGenomics and Informatics Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Hope Center for Neurological Disorders, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neurology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Division of Biostatistics, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA USA
- Neuroscience Research Australia, Sydney, Australia
- School of Medical Sciences, University of New South Wales, Sydney, Australia
- Department of Neurology, Keck School of Medicine of University of Southern California, Los Angeles, CA USA
- Department of Genetics, Washington University in Saint Louis School of Medicine, Saint Louis, MO USA
| |
Collapse
|
113
|
Ferrara SJ, Chaudhary P, DeBell MJ, Marracci G, Miller H, Calkins E, Pocius E, Napier BA, Emery B, Bourdette D, Scanlan TS. TREM2 is thyroid hormone regulated making the TREM2 pathway druggable with ligands for thyroid hormone receptor. Cell Chem Biol 2022; 29:239-248.e4. [PMID: 34375614 PMCID: PMC8818810 DOI: 10.1016/j.chembiol.2021.07.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/03/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022]
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) is a cell surface receptor on macrophages and microglia that senses and responds to disease-associated signals to regulate the phenotype of these innate immune cells. The TREM2 signaling pathway has been implicated in a variety of diseases ranging from neurodegeneration in the central nervous system to metabolic disease in the periphery. Here, we report that TREM2 is a thyroid hormone-regulated gene and its expression in macrophages and microglia is stimulated by thyroid hormone and synthetic thyroid hormone agonists (thyromimetics). Our findings report the endocrine regulation of TREM2 by thyroid hormone, and provide a unique opportunity to drug the TREM2 signaling pathway with orally active small-molecule therapeutic agents.
Collapse
MESH Headings
- Acetates/chemical synthesis
- Acetates/pharmacology
- Animals
- Binding Sites
- Brain/drug effects
- Brain/immunology
- Brain/pathology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Regulation
- Humans
- Immunity, Innate
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/pathology
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/immunology
- Microglia/pathology
- Models, Molecular
- Phenols/chemical synthesis
- Phenols/pharmacology
- Phenoxyacetates/pharmacology
- Promoter Regions, Genetic
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- Protein Interaction Domains and Motifs
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Response Elements
- Retinoid X Receptors/chemistry
- Retinoid X Receptors/genetics
- Retinoid X Receptors/metabolism
- Signal Transduction
- Thyroid Hormones/pharmacology
Collapse
Affiliation(s)
- Skylar J Ferrara
- Department of Chemical Physiology and Biochemistry and Program in Chemical Biology, Oregon Health & Science University, L334, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Priya Chaudhary
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Margaret J DeBell
- Department of Chemical Physiology and Biochemistry and Program in Chemical Biology, Oregon Health & Science University, L334, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Gail Marracci
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Hannah Miller
- Department of Chemical Physiology and Biochemistry and Program in Chemical Biology, Oregon Health & Science University, L334, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Evan Calkins
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Edvinas Pocius
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Brooke A Napier
- Department of Biology, Portland State University, OR 97201, USA
| | - Ben Emery
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; Jungers Center for Neurosciences Research, Oregon Health & Science University, Portland, OR 97239, USA
| | - Dennis Bourdette
- VA Portland Health Care System, Portland, OR 97239, USA; Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Thomas S Scanlan
- Department of Chemical Physiology and Biochemistry and Program in Chemical Biology, Oregon Health & Science University, L334, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
114
|
Reifschneider A, Robinson S, van Lengerich B, Gnörich J, Logan T, Heindl S, Vogt MA, Weidinger E, Riedl L, Wind K, Zatcepin A, Pesämaa I, Haberl S, Nuscher B, Kleinberger G, Klimmt J, Götzl JK, Liesz A, Bürger K, Brendel M, Levin J, Diehl‐Schmid J, Suh J, Di Paolo G, Lewcock JW, Monroe KM, Paquet D, Capell A, Haass C. Loss of TREM2 rescues hyperactivation of microglia, but not lysosomal deficits and neurotoxicity in models of progranulin deficiency. EMBO J 2022; 41:e109108. [PMID: 35019161 PMCID: PMC8844989 DOI: 10.15252/embj.2021109108] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Haploinsufficiency of the progranulin (PGRN)-encoding gene (GRN) causes frontotemporal lobar degeneration (GRN-FTLD) and results in microglial hyperactivation, TREM2 activation, lysosomal dysfunction, and TDP-43 deposition. To understand the contribution of microglial hyperactivation to pathology, we used genetic and pharmacological approaches to suppress TREM2-dependent transition of microglia from a homeostatic to a disease-associated state. Trem2 deficiency in Grn KO mice reduced microglia hyperactivation. To explore antibody-mediated pharmacological modulation of TREM2-dependent microglial states, we identified antagonistic TREM2 antibodies. Treatment of macrophages from GRN-FTLD patients with these antibodies led to reduced TREM2 signaling due to its enhanced shedding. Furthermore, TREM2 antibody-treated PGRN-deficient microglia derived from human-induced pluripotent stem cells showed reduced microglial hyperactivation, TREM2 signaling, and phagocytic activity, but lysosomal dysfunction was not rescued. Similarly, lysosomal dysfunction, lipid dysregulation, and glucose hypometabolism of Grn KO mice were not rescued by TREM2 ablation. Synaptic loss and neurofilament light-chain (NfL) levels, a biomarker for neurodegeneration, were further elevated in the Grn/Trem2 KO cerebrospinal fluid (CSF). These findings suggest that TREM2-dependent microglia hyperactivation in models of GRN deficiency does not promote neurotoxicity, but rather neuroprotection.
Collapse
Affiliation(s)
- Anika Reifschneider
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Sophie Robinson
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
| | | | - Johannes Gnörich
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Todd Logan
- Denali Therapeutics Inc.South San FranciscoCAUSA
| | - Steffanie Heindl
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | | | - Endy Weidinger
- Department of NeurologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Lina Riedl
- Department of Psychiatry and PsychotherapySchool of MedicineTechnical University of MunichMunichGermany
| | - Karin Wind
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Artem Zatcepin
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Ida Pesämaa
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
| | | | - Brigitte Nuscher
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | | | - Julien Klimmt
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Julia K Götzl
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Arthur Liesz
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Katharina Bürger
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Department of NeurologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Janine Diehl‐Schmid
- Department of NeurologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Department of Psychiatry and PsychotherapySchool of MedicineTechnical University of MunichMunichGermany
| | - Jung Suh
- Denali Therapeutics Inc.South San FranciscoCAUSA
| | | | | | | | - Dominik Paquet
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Anja Capell
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christian Haass
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| |
Collapse
|
115
|
Soluble TREM2 inhibits secondary nucleation of Aβ fibrillization and enhances cellular uptake of fibrillar Aβ. Proc Natl Acad Sci U S A 2022; 119:2114486119. [PMID: 35082148 PMCID: PMC8812518 DOI: 10.1073/pnas.2114486119] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 01/21/2023] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is a single-pass transmembrane receptor of the immunoglobulin superfamily that is secreted in a soluble (sTREM2) form. Mutations in TREM2 have been linked to increased risk of Alzheimer's disease (AD). A prominent neuropathological component of AD is deposition of the amyloid-β (Aβ) into plaques, particularly Aβ40 and Aβ42. While the membrane-bound form of TREM2 is known to facilitate uptake of Aβ fibrils and the polarization of microglial processes toward amyloid plaques, the role of its soluble ectodomain, particularly in interactions with monomeric or fibrillar Aβ, has been less clear. Our results demonstrate that sTREM2 does not bind to monomeric Aβ40 and Aβ42, even at a high micromolar concentration, while it does bind to fibrillar Aβ42 and Aβ40 with equal affinities (2.6 ± 0.3 µM and 2.3 ± 0.4 µM). Kinetic analysis shows that sTREM2 inhibits the secondary nucleation step in the fibrillization of Aβ, while having little effect on the primary nucleation pathway. Furthermore, binding of sTREM2 to fibrils markedly enhanced uptake of fibrils into human microglial and neuroglioma derived cell lines. The disease-associated sTREM2 mutant, R47H, displayed little to no effect on fibril nucleation and binding, but it decreased uptake and functional responses markedly. We also probed the structure of the WT sTREM2-Aβ fibril complex using integrative molecular modeling based primarily on the cross-linking mass spectrometry data. The model shows that sTREM2 binds fibrils along one face of the structure, leaving a second, mutation-sensitive site free to mediate cellular binding and uptake.
Collapse
|
116
|
Brown GC, St George-Hyslop P. Does Soluble TREM2 Protect Against Alzheimer's Disease? Front Aging Neurosci 2022; 13:834697. [PMID: 35153729 PMCID: PMC8831327 DOI: 10.3389/fnagi.2021.834697] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/31/2021] [Indexed: 02/02/2023] Open
Abstract
Triggering Receptor Expressed in Myeloid Cells 2 (TREM2) is a pattern recognition receptor on myeloid cells, and is upregulated on microglia surrounding amyloid plaques in Alzheimer's disease (AD). Rare, heterozygous mutations in TREM2 (e.g., R47H) increase AD risk several fold. TREM2 can be cleaved at the plasma membrane by metalloproteases to release the ectodomain as soluble TREM2 (sTREM2). Wild-type sTREM2 binds oligomeric amyloid beta (Aβ) and acts as an extracellular chaperone, blocking and reversing Aβ oligomerization and fibrillization, and preventing Aβ-induced neuronal loss in vitro. Whereas, R47H sTREM2 increases Aβ fibrillization and neurotoxicity. AD brains expressing R47H TREM2 have more fibrous plaques with more neuritic pathology around these plaques, consistent with R47H sTREM2 promoting Aβ fibrillization relative to WT sTREM2. Brain expression or injection of wild-type sTREM2 reduces pathology in amyloid models of AD in mice, indicating that wild-type sTREM2 is protective against amyloid pathology. Levels of sTREM2 in cerebrospinal fluid (CSF) fall prior to AD, rise in early AD, and fall again in late AD. People with higher sTREM2 levels in CSF progress more slowly into and through AD than do people with lower sTREM2 levels, suggesting that sTREM2 protects against AD. However, some of these experiments can be interpreted as full-length TREM2 protecting rather than sTREM2, and to distinguish between these two possibilities, we need more experiments testing whether sTREM2 itself protects in AD and AD models, and at what stage of disease. If sTREM2 is protective, then treatments could be designed to elevate sTREM2 in AD.
Collapse
Affiliation(s)
- Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom,*Correspondence: Guy C. Brown
| | - Peter St George-Hyslop
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
117
|
Emilsson V, Gudmundsdottir V, Gudjonsson A, Jonmundsson T, Jonsson BG, Karim MA, Ilkov M, Staley JR, Gudmundsson EF, Launer LJ, Lindeman JH, Morton NM, Aspelund T, Lamb JR, Jennings LL, Gudnason V. Coding and regulatory variants are associated with serum protein levels and disease. Nat Commun 2022; 13:481. [PMID: 35079000 PMCID: PMC8789809 DOI: 10.1038/s41467-022-28081-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 01/07/2022] [Indexed: 12/20/2022] Open
Abstract
Circulating proteins can be used to diagnose and predict disease-related outcomes. A deep serum proteome survey recently revealed close associations between serum protein networks and common disease. In the current study, 54,469 low-frequency and common exome-array variants were compared to 4782 protein measurements in the serum of 5343 individuals from the AGES Reykjavik cohort. This analysis identifies a large number of serum proteins with genetic signatures overlapping those of many diseases. More specifically, using a study-wide significance threshold, we find that 2021 independent exome array variants are associated with serum levels of 1942 proteins. These variants reside in genetic loci shared by hundreds of complex disease traits, highlighting serum proteins' emerging role as biomarkers and potential causative agents of a wide range of diseases.
Collapse
Affiliation(s)
- Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Reykjavík, Iceland.
| | | | | | | | | | - Mohd A Karim
- Wellcome Trust Sanger Institute, Welcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| | - Marjan Ilkov
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Kopavogur, Iceland
| | - James R Staley
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Elias F Gudmundsson
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Kopavogur, Iceland
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, 20892-9205, USA
| | - Jan H Lindeman
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Nicholas M Morton
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Kopavogur, Iceland
| | - John R Lamb
- GNF Novartis, 10675 John Jay Hopkins Drive, San Diego, CA, 92121, USA
| | - Lori L Jennings
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA, 02139, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, IS-201 Kopavogur, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Reykjavík, Iceland.
| |
Collapse
|
118
|
Filipello F, Goldsbury C, Feng YS, Locca A, Karch CM, Piccio L. Soluble TREM2: Innocent bystander or active player in neurological diseases? Neurobiol Dis 2022; 165:105630. [PMID: 35041990 PMCID: PMC10108835 DOI: 10.1016/j.nbd.2022.105630] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/14/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor expressed by macrophages and microglia in the central nervous system (CNS). TREM2 has attracted a lot of interest in the past decade for its critical role in modulating microglia functions under homeostatic conditions and in neurodegenerative diseases. Genetic variation in TREM2 is sufficient to cause Nasu-Hakola disease, a rare pre-senile dementia with bone cysts, and to increase risk for Alzheimer's disease, frontotemporal dementia, and other neurodegenerative disorders. Beyond the role played by TREM2 genetic variants in these diseases, TREM2 engagement is a key step in microglia activation in response to different types of tissue injury (e.g. β-Amyloid deposition, demyelination, apoptotic cell death) leading to enhanced microglia metabolism, phagocytosis, proliferation and survival. TREM2 also exists as a soluble form (sTREM2), generated from receptor shedding or alternative splicing, which is detectable in plasma and cerebrospinal fluid (CSF). Genetic variation, physiological conditions and disease status impact CSF sTREM2 levels. Clinical and preclinical studies suggest that targeting and/or monitoring sTREM2 could have clinical and therapeutic implications. Despite the critical role of sTREM2 in neurologic disease, its function remains poorly understood. Here, we review the current literature on sTREM2 regarding its origin, genetic variation, and possible functions as a biomarker in neurological disorders and as a potential active player in CNS diseases and target for therapies.
Collapse
Affiliation(s)
- Fabia Filipello
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Claire Goldsbury
- Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
| | - You Shih Feng
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Alberto Locca
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
119
|
Timsina J, Gomez-Fonseca D, Wang L, Do A, Western D, Alvarez I, Aguilar M, Pastor P, Henson RL, Herries E, Xiong C, Schindler SE, Fagan AM, Bateman RJ, Farlow M, Morris JC, Perrin R, Moulder K, Hassenstab J, Chhatwal J, Mori H, Sung YJ, Cruchaga C. Comparative Analysis of Alzheimer's Disease Cerebrospinal Fluid Biomarkers Measurement by Multiplex SOMAscan Platform and Immunoassay-Based Approach. J Alzheimers Dis 2022; 89:193-207. [PMID: 35871346 PMCID: PMC9562128 DOI: 10.3233/jad-220399] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND The SOMAscan assay has an advantage over immunoassay-based methods because it measures a large number of proteins in a cost-effective manner. However, the performance of this technology compared to the routinely used immunoassay techniques needs to be evaluated. OBJECTIVE We performed comparative analyses of SOMAscan and immunoassay-based protein measurements for five cerebrospinal fluid (CSF) proteins associated with Alzheimer's disease (AD) and neurodegeneration: NfL, Neurogranin, sTREM2, VILIP-1, and SNAP-25. METHODS We compared biomarkers measured in ADNI (N = 689), Knight-ADRC (N = 870), DIAN (N = 115), and Barcelona-1 (N = 92) cohorts. Raw protein values were transformed using z-score in order to combine measures from the different studies. sTREM2 and VILIP-1 had more than one analyte in SOMAscan; all available analytes were evaluated. Pearson's correlation coefficients between SOMAscan and immunoassays were calculated. Receiver operating characteristic curve and area under the curve were used to compare prediction accuracy of these biomarkers between the two platforms. RESULTS Neurogranin, VILIP-1, and NfL showed high correlation between SOMAscan and immunoassay measures (r > 0.9). sTREM2 had a fair correlation (r > 0.6), whereas SNAP-25 showed weak correlation (r = 0.06). Measures in both platforms provided similar predicted performance for all biomarkers except SNAP-25 and one of the sTREM2 analytes. sTREM2 showed higher AUC for SOMAscan based measures. CONCLUSION Our data indicate that SOMAscan performs as well as immunoassay approaches for NfL, Neurogranin, VILIP-1, and sTREM2. Our study shows promise for using SOMAscan as an alternative to traditional immunoassay-based measures. Follow-up investigation will be required for SNAP-25 and additional established biomarkers.
Collapse
Affiliation(s)
- Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Duber Gomez-Fonseca
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Anh Do
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Dan Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ignacio Alvarez
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Pau Pastor
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Rachel L. Henson
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Elizabeth Herries
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E. Schindler
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M. Fagan
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Martin Farlow
- Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Health, Indianapolis, IN, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Krista Moulder
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hiroshi Mori
- Dept. of Clinical Neuroscience, Osaka City University Medical School, Nagaoka Sutoku University, Japan
| | | | | | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurologic Diseases, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
120
|
Tanaka M, Yamakage H, Muranaka K, Yamada T, Araki R, Ogo A, Matoba Y, Watanabe T, Saito M, Kurita S, Yonezawa K, Tanaka T, Suzuki M, Sawamura M, Matsumoto M, Nishimura M, Kusakabe T, Wada H, Hasegawa K, Kotani K, Noda M, Satoh-Asahara N. Higher Serum Soluble TREM2 as a Potential Indicative Biomarker for Cognitive Impairment in Inadequately Controlled Type 2 Diabetes Without Obesity: The DOR-KyotoJ-1. Front Endocrinol (Lausanne) 2022; 13:880148. [PMID: 35592778 PMCID: PMC9110765 DOI: 10.3389/fendo.2022.880148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Type 2 diabetes is a risk factor for dementia. We investigated whether serum levels of soluble triggering receptor expressed on myeloid cell 2 (sTREM2), a soluble form of the cell surface receptor TREM2, were predictive of cognitive impairment in type 2 diabetes without obesity. METHODS A total of 166 Japanese patients with type 2 diabetes without obesity were followed-up for 2 years. We measured clinical parameters, assessed cognitive function using the mini-mental state examination (MMSE), quantified and divided serum sTREM2 levels into quartiles, and examined the longitudinal associations. RESULTS During the follow-up, HbA1c levels were elevated in 98 patients and decreased in 68 patients. In the HbA1c-elevated group, higher sTREM2 levels at baseline showed a significant association with a greater tendency for reduction in MMSE scores (P for trend = 0.015), whereas they were not significantly associated with other examined parameters. In the HbA1c-decreased group, there was no significant association between sTREM2 levels at baseline and changes in MMSE scores, but higher sTREM2 levels at baseline were significantly associated with a greater tendency for reduction in waist circumference (P for trend = 0.027), homeostasis model assessment of insulin resistance (P for trend = 0.039), and sTREM2 levels (P for trend = 0.023). CONCLUSIONS Glycemic control is suggested to be important in preventing cognitive impairment in patients with type 2 diabetes without obesity. Higher serum sTREM2 levels would be a predictive marker for cognitive impairment in inadequately controlled type 2 diabetes without obesity.
Collapse
Affiliation(s)
- Masashi Tanaka
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
- Department of Physical Therapy, Health Science University, Yamanashi, Japan
- *Correspondence: Noriko Satoh-Asahara, ; Masashi Tanaka,
| | - Hajime Yamakage
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kazuya Muranaka
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Tsutomu Yamada
- Department of Endocrinology and Diabetes, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Rika Araki
- Department of Diabetes and Endocrinology, National Hospital Organization National Mie Hospital, Mie, Japan
| | - Atsushi Ogo
- Department of Metabolism and Endocrinology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Yuka Matoba
- Department of Metabolism and Endocrinology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Tetsuhiro Watanabe
- Department of Metabolism and Endocrinology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Miho Saito
- Department of Internal Medicine, Tokushima National Hospital, Tokushima, Japan
| | - Seiichiro Kurita
- Department of Internal Medicine, National Hospital Organization Kanazawa Medical Center, Ishikawa, Japan
| | - Kazuya Yonezawa
- Department of Clinical Research, Hakodate National Hospital, Hokkaido, Japan
| | - Tsuyoshi Tanaka
- Department of Endocrinology and Metabolism, National Hospital Organization Mie Chuo Medical Center, Mie, Japan
| | - Masahiro Suzuki
- Department of Clinical Research, National Hospital Organization Saitama Hospital, Saitama, Japan
| | - Morio Sawamura
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Gunma, Japan
| | - Morio Matsumoto
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Gunma, Japan
| | - Motonobu Nishimura
- Department of Diabetes and Endocrinology, National Hospital Organization Chibahigashi National Hospital, Chiba, Japan
| | - Toru Kusakabe
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Hiromichi Wada
- Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Koji Hasegawa
- Division of Translational Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kazuhiko Kotani
- Center for Community Medicine, Jichi Medical University, Tochigi, Japan
| | - Mitsuhiko Noda
- Department of Diabetes, Metabolism and Endocrinology, Ichikawa Hospital, International University of Health and Welfare, Chiba, Japan
- Department of Endocrinology and Diabetes, Saitama Medical University, Saitama, Japan
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
- Department of Metabolic Syndrome and Nutritional Science, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
- *Correspondence: Noriko Satoh-Asahara, ; Masashi Tanaka,
| |
Collapse
|
121
|
Galectin-3 is elevated in CSF and is associated with Aβ deposits and tau aggregates in brain tissue in Alzheimer's disease. Acta Neuropathol 2022; 144:843-859. [PMID: 35895141 PMCID: PMC9547798 DOI: 10.1007/s00401-022-02469-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 01/26/2023]
Abstract
Galectin-3 (Gal-3) is a beta-galactosidase binding protein involved in microglial activation in the central nervous system (CNS). We previously demonstrated the crucial deleterious role of Gal-3 in microglial activation in Alzheimer's disease (AD). Under AD conditions, Gal-3 is primarily expressed by microglial cells clustered around Aβ plaques in both human and mouse brain, and knocking out Gal-3 reduces AD pathology in AD-model mice. To further unravel the importance of Gal-3-associated inflammation in AD, we aimed to investigate the Gal-3 inflammatory response in the AD continuum. First, we measured Gal-3 levels in neocortical and hippocampal tissue from early-onset AD patients, including genetic and sporadic cases. We found that Gal-3 levels were significantly higher in both cortex and hippocampus in AD subjects. Immunohistochemistry revealed that Gal-3+ microglial cells were associated with amyloid plaques of a larger size and more irregular shape and with neurons containing tau-inclusions. We then analyzed the levels of Gal-3 in cerebrospinal fluid (CSF) from AD patients (n = 119) compared to control individuals (n = 36). CSF Gal-3 levels were elevated in AD patients compared to controls and more strongly correlated with tau (p-Tau181 and t-tau) and synaptic markers (GAP-43 and neurogranin) than with amyloid-β. Lastly, principal component analysis (PCA) of AD biomarkers revealed that CSF Gal-3 clustered and associated with other CSF neuroinflammatory markers, including sTREM-2, GFAP, and YKL-40. This neuroinflammatory component was more highly expressed in the CSF from amyloid-β positive (A+), CSF p-Tau181 positive (T+), and biomarker neurodegeneration positive/negative (N+/-) (A + T + N+/-) groups compared to the A + T-N- group. Overall, Gal-3 stands out as a key pathological biomarker of AD pathology that is measurable in CSF and, therefore, a potential target for disease-modifying therapies involving the neuroinflammatory response.
Collapse
|
122
|
Brosseron F, Maass A, Kleineidam L, Ravichandran KA, González PG, McManus RM, Ising C, Santarelli F, Kolbe CC, Häsler LM, Wolfsgruber S, Marquié M, Boada M, Orellana A, de Rojas I, Röske S, Peters O, Cosma NC, Cetindag A, Wang X, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Schott BH, Bürger K, Janowitz D, Dichgans M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Göerß D, Laske C, Munk MH, Düzel E, Yakupov R, Dobisch L, Metzger CD, Glanz W, Ewers M, Dechent P, Haynes JD, Scheffler K, Roy N, Rostamzadeh A, Teunissen CE, Marchant NL, Spottke A, Jucker M, Latz E, Wagner M, Mengel D, Synofzik M, Jessen F, Ramirez A, Ruiz A, Heneka MT. Soluble TAM receptors sAXL and sTyro3 predict structural and functional protection in Alzheimer's disease. Neuron 2021; 110:1009-1022.e4. [PMID: 34995486 DOI: 10.1016/j.neuron.2021.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 11/17/2021] [Accepted: 12/10/2021] [Indexed: 10/19/2022]
Abstract
There is an urgent need to improve the understanding of neuroinflammation in Alzheimer's disease (AD). We analyzed cerebrospinal fluid inflammatory biomarker correlations to brain structural volume and longitudinal cognitive outcomes in the DELCODE study and in a validation cohort of the F.ACE Alzheimer Center Barcelona. We investigated whether respective biomarker changes are evident before onset of cognitive impairment. YKL-40; sTREM2; sAXL; sTyro3; MIF; complement factors C1q, C4, and H; ferritin; and ApoE protein were elevated in pre-dementia subjects with pathological levels of tau or other neurodegeneration markers, demonstrating tight interactions between inflammation and accumulating neurodegeneration even before onset of symptoms. Intriguingly, higher levels of ApoE and soluble TAM receptors sAXL and sTyro3 were related to larger brain structure and stable cognitive outcome at follow-up. Our findings indicate a protective mechanism relevant for intervention strategies aiming to regulate neuroinflammation in subjects with no or subjective symptoms but underlying AD pathology profile.
Collapse
Affiliation(s)
- Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kishore Aravind Ravichandran
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Pablo García González
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christina Ising
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Francesco Santarelli
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Carl-Christian Kolbe
- University of Bonn Medical Center, Institute of Innate Immune, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Lisa M Häsler
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Marta Marquié
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Röske
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicoleta-Carmen Cosma
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Arda Cetindag
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Xiao Wang
- Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany; Department of Psychiatry and Psychotherapy, Technical University Munich, 81675 Munich, Germany
| | - Eike J Spruth
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), Charitéplatz 1, Berlin 10117, Germany; Department of Psychiatry and Psychotherapy, Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, Göttingen 37075, Germany; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H Schott
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, Göttingen 37075, Germany; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, University of Göttingen, Von-Siebold-Str. 5, 37075 Göttingen, Germany; Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Katharina Bürger
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany; Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany; Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany; Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, Rostock 18147, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Gehlsheimer Str. 20, Rostock 18147, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Doreen Göerß
- Department of Psychosomatic Medicine, Rostock University Medical Center, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Renat Yakupov
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Coraline D Metzger
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany; Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Leipziger Straße 44, Magdeburg 39120, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, Munich 81377, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - John Dylan Haynes
- Bernstein Center for Computational Neurosciences, Charité - Universitätsmedizin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical centers, Vrije Universiteit, Amsterdam, the Netherlands
| | | | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Mathias Jucker
- Hertie Institute for Clinical Brain Research, Department of Cellular Neurology, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; University of Bonn Medical Center, Institute of Innate Immune, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany
| | - David Mengel
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 27, Tübingen 72076, Germany; Division Translational Genomics of Neurodegenerative Diseases, Center for Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076 Tübingen, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany; Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Köln, Germany; Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundacío ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Bonn 53127, Germany; Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Venusberg-Campus 1, 53127 Bonn, Germany; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, 4362 Esch-sur- Alzette, Luxembourg; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, 55 Lake Avenue, North Worcester, Massachusetts 01655, USA.
| | | |
Collapse
|
123
|
Kwak K, Giovanello KS, Bozoki A, Styner M, Dayan E. Subtyping of mild cognitive impairment using a deep learning model based on brain atrophy patterns. Cell Rep Med 2021; 2:100467. [PMID: 35028609 PMCID: PMC8714856 DOI: 10.1016/j.xcrm.2021.100467] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/08/2021] [Accepted: 11/13/2021] [Indexed: 12/28/2022]
Abstract
Trajectories of cognitive decline vary considerably among individuals with mild cognitive impairment (MCI). To address this heterogeneity, subtyping approaches have been developed, with the objective of identifying more homogeneous subgroups. To date, subtyping of MCI has been based primarily on cognitive measures, often resulting in indistinct boundaries between subgroups and limited validity. Here, we introduce a subtyping method for MCI based solely upon brain atrophy. We train a deep learning model to differentiate between Alzheimer's disease (AD) and cognitively normal (CN) subjects based on whole-brain MRI features. We then deploy the trained model to classify MCI subjects based on whole-brain gray matter resemblance to AD-like or CN-like patterns. We subsequently validate the subtyping approach using cognitive, clinical, fluid biomarker, and molecular imaging data. Overall, the results suggest that atrophy patterns in MCI are sufficiently heterogeneous and can thus be used to subtype individuals into biologically and clinically meaningful subgroups.
Collapse
Affiliation(s)
- Kichang Kwak
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kelly S. Giovanello
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrea Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Martin Styner
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eran Dayan
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
124
|
Pereira JB, Janelidze S, Smith R, Mattsson-Carlgren N, Palmqvist S, Teunissen CE, Zetterberg H, Stomrud E, Ashton NJ, Blennow K, Hansson O. Plasma GFAP is an early marker of amyloid-β but not tau pathology in Alzheimer's disease. Brain 2021; 144:3505-3516. [PMID: 34259835 PMCID: PMC8677538 DOI: 10.1093/brain/awab223] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/24/2021] [Accepted: 06/05/2021] [Indexed: 11/13/2022] Open
Abstract
Although recent clinical trials targeting amyloid-β in Alzheimer's disease have shown promising results, there is increasing evidence suggesting that understanding alternative disease pathways that interact with amyloid-β metabolism and amyloid pathology might be important to halt the clinical deterioration. In particular, there is evidence supporting a critical role of astroglial activation and astrocytosis in Alzheimer's disease. However, so far, no studies have assessed whether astrocytosis is independently related to either amyloid-β or tau pathology in vivo. To address this question, we determined the levels of the astrocytic marker GFAP in plasma and CSF of 217 amyloid-β-negative cognitively unimpaired individuals, 71 amyloid-β-positive cognitively unimpaired individuals, 78 amyloid-β-positive cognitively impaired individuals, 63 amyloid-β-negative cognitively impaired individuals and 75 patients with a non-Alzheimer's disease neurodegenerative disorder from the Swedish BioFINDER-2 study. Participants underwent longitudinal amyloid-β (18F-flutemetamol) and tau (18F-RO948) PET as well as cognitive testing. We found that plasma GFAP concentration was significantly increased in all amyloid-β-positive groups compared with participants without amyloid-β pathology (P < 0.01). In addition, there were significant associations between plasma GFAP with higher amyloid-β-PET signal in all amyloid-β-positive groups, but also in cognitively normal individuals with normal amyloid-β values (P < 0.001), which remained significant after controlling for tau-PET signal. Furthermore, plasma GFAP could predict amyloid-β-PET positivity with an area under the curve of 0.76, which was greater than the performance achieved by CSF GFAP (0.69) and other glial markers (CSF YKL-40: 0.64, soluble TREM2: 0.71). Although correlations were also observed between tau-PET and plasma GFAP, these were no longer significant after controlling for amyloid-β-PET. In contrast to plasma GFAP, CSF GFAP concentration was significantly increased in non-Alzheimer's disease patients compared to other groups (P < 0.05) and correlated with amyloid-β-PET only in amyloid-β-positive cognitively impaired individuals (P = 0.005). Finally, plasma GFAP was associated with both longitudinal amyloid-β-PET and cognitive decline, and mediated the effect of amyloid-β-PET on tau-PET burden, suggesting that astrocytosis secondary to amyloid-β aggregation might promote tau accumulation. Altogether, these findings indicate that plasma GFAP is an early marker associated with brain amyloid-β pathology but not tau aggregation, even in cognitively normal individuals with a normal amyloid-β status. This suggests that plasma GFAP should be incorporated in current hypothetical models of Alzheimer's disease pathogenesis and be used as a non-invasive and accessible tool to detect early astrocytosis secondary to amyloid-β pathology.
Collapse
Affiliation(s)
- Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Nicholas J Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
125
|
Teipel SJ, Dyrba M, Ballarini T, Brosseron F, Bruno D, Buerger K, Cosma NC, Dechent P, Dobisch L, Düzel E, Ewers M, Fliessbach K, Haynes JD, Janowitz D, Kilimann I, Laske C, Maier F, Metzger CD, Munk MH, Peters O, Pomara N, Preis L, Priller J, Ramírez A, Roy N, Scheffler K, Schneider A, Schott BH, Spottke A, Spruth EJ, Wagner M, Wiltfang J, Jessen F, Heneka MT. Association of Cholinergic Basal Forebrain Volume and Functional Connectivity with Markers of Inflammatory Response in the Alzheimer’s Disease Spectrum. J Alzheimers Dis 2021; 85:1267-1282. [DOI: 10.3233/jad-215196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Inflammation has been described as a key pathogenic event In Alzheimer’s disease (AD), downstream of amyloid and tau pathology. Preclinical and clinical data suggest that the cholinergic basal forebrain may moderate inflammatory response to different pathologies. Objective: To study the association of cholinergic basal forebrain volume and functional connectivity with measures of neuroinflammation in people from the AD spectrum. Methods: We studied 261 cases from the DELCODE cohort, including people with subjective cognitive decline, mild cognitive impairment, AD dementia, first degree relatives, and healthy controls. Using Bayesian ANCOVA, we tested associations of MRI indices of cholinergic basal forebrain volume and functional connectivity with cerebrospinal fluid (CSF) levels of sTREM2 as a marker of microglia activation, and serum levels of complement C3. Using Bayesian elastic net regression, we determined associations between basal forebrain measures and a large inflammation marker panel from CSF and serum. Results: We found anecdotal to moderate evidence in favor of the absence of an effect of basal forebrain volume and functional connectivity on CSF sTREM2 and serum C3 levels both in Aβ 42/ptau-positive and negative cases. Bayesian elastic net regression identified several CSF and serum markers of inflammation that were associated with basal forebrain volume and functional connectivity. The effect sizes were moderate to small. Conclusion: Our data-driven analyses generate the hypothesis that cholinergic basal forebrain may be involved in the neuroinflammation response to Aβ 42 and phospho-tau pathology in people from the AD spectrum. This hypothesis needs to be tested in independent samples.
Collapse
Affiliation(s)
- Stefan J. Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Martin Dyrba
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | | | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Davide Bruno
- School of Psychology, Liverpool John Moores University, Liverpool, UK
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Nicoleta-Carmen Cosma
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Peter Dechent
- MR-Research in Neurosciences, Georg-August-University, Goettingen, Germany
| | - Laura Dobisch
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - John D. Haynes
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research, Tuebingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tuebingen, Germany
| | - Franziska Maier
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Coraline D. Metzger
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
| | - Matthias H. Munk
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research, Tuebingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Tuebingen, Germany
| | - Oliver Peters
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Nunzio Pomara
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, School of Medicine, New York University, New York City, NY, USA
| | - Lukas Preis
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité –Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Alfredo Ramírez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Nina Roy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tuebingen, Tuebingen, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Björn H. Schott
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Goettingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Eike J. Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité –Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Michael T. Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department for Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
126
|
Yamagishi S, Iga Y, Ikegaya S, Kakiuchi T, Ohba H, Nishiyama S, Fukomoto D, Kanazawa M, Harada N, Tsukada H, Sato K, Ouchi Y. In vivo alterations of mitochondrial activity and amyloidosis in early-stage senescence-accelerated mice: a positron emission tomography study. J Neuroinflammation 2021; 18:288. [PMID: 34893067 PMCID: PMC8665644 DOI: 10.1186/s12974-021-02343-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/05/2021] [Indexed: 11/10/2022] Open
Abstract
Purpose While marked reductions in neural activity and mitochondrial function have been reported in Alzheimer’s disease (AD), the degree of mitochondrial activity in mild cognitive impairment (MCI) or early-stage AD remains unexplored. Here, we used positron emission tomography (PET) to examine the direct relationship between mitochondrial activity (18F-BCPP-EF) and β-amyloid (Aβ) deposition (11C-PiB) in the same brains of senescence-accelerated mouse prone 10 (SAMP10) mice, an Aβ-developing neuroinflammatory animal model showing accelerated senescence with deterioration in cognitive functioning similar to that in MCI. Methods Five- to 25-week-old SAMP10 and control SAMR1 mice, were used in the experiments. PET was used to measure the binding levels (standard uptake value ratios; SUVRs) of [18F]2-tert-butyl-4-chloro-5-2H-pyridazin-3-one (18F-BCPP-EF) for mitochondrial complex 1 availability, and 11C-PiB for Aβ deposition, in the same animals, and immunohistochemistry for ATPB (an ATP synthase on the mitochondrial inner membrane) was also performed, to determine changes in mitochondrial activity in relation to amyloid burden during the early stage of cognitive impairment. Results The SUVR of 18F-BCPP-EF was significantly lower and that of 11C-PiB was higher in the 15-week-old SAMP10 mice than in the control and 5-week-old SAMP10 mice. The two parameters were found to negatively correlate with each other. The immunohistochemical analysis demonstrated temporal upregulation of ATPB levels at 15-week-old, but decreased at 25 week-old SAMP10 mice. Conclusion The present results provide in vivo evidence of a decrease in mitochondrial energy production and elevated amyloidosis at an early stage in SAMP10 mice. The inverse correlation between these two phenomena suggests a concurrent change in neuronal energy failure by Aβ-induced elevation of neuroinflammatory responses. Comparison of PET data with histological findings suggests that temporal increase of ATPB level may not be neurofunctionally implicated during neuropathological processes, including Aβ pathology, in an animal model of early-phase AD spectrum disorder. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02343-4.
Collapse
Affiliation(s)
- Satoru Yamagishi
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yurika Iga
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shunsuke Ikegaya
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takeharu Kakiuchi
- Central Research Laboratory, Hamamatsu Photonics KK, Hamamatsu, Japan
| | - Hiroyuki Ohba
- Central Research Laboratory, Hamamatsu Photonics KK, Hamamatsu, Japan
| | - Shingo Nishiyama
- Central Research Laboratory, Hamamatsu Photonics KK, Hamamatsu, Japan
| | - Daisuke Fukomoto
- Central Research Laboratory, Hamamatsu Photonics KK, Hamamatsu, Japan
| | | | - Norihiro Harada
- Central Research Laboratory, Hamamatsu Photonics KK, Hamamatsu, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics KK, Hamamatsu, Japan
| | - Kohji Sato
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
127
|
Sheng X, Yao Y, Huang R, Xu Y, Zhu Y, Chen L, Zhang L, Wang W, Zhuo R, Can D, Chang CF, Zhang YW, Xu H, Bu G, Zhong L, Chen XF. Identification of the minimal active soluble TREM2 sequence for modulating microglial phenotypes and amyloid pathology. J Neuroinflammation 2021; 18:286. [PMID: 34893068 PMCID: PMC8665564 DOI: 10.1186/s12974-021-02340-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022] Open
Abstract
Background TREM2 is a microglial receptor genetically linked to the risk for Alzheimer’s disease (AD). The cerebrospinal fluid (CSF) levels of soluble TREM2 (sTREM2) have emerged as a valuable biomarker for the disease progression in AD and higher CSF levels of sTREM2 are linked to slower cognitive decline. Increasing sTREM2 in mouse models of amyloidosis reduces amyloid-related pathology through modulating microglial functions, suggesting a beneficial role of sTREM2 in microglia biology and AD pathology. Methods In the current study, we performed serial C- and N-terminal truncations of sTREM2 protein to define the minimal sequence requirement for sTREM2 function. We initially assessed the impacts of sTREM2 mutants on microglial functions by measuring cell viability and inflammatory responses. The binding of the sTREM2 mutants to oligomeric Aβ was determined by solid-phase protein binding assay and dot blot assay. We further evaluated the impacts of sTREM2 mutants on amyloid-related pathology by direct stereotaxic injection of sTREM2 proteins into the brain of 5xFAD mice. Results We found that both sTREM2 fragments 41–81 and 51–81 enhance cell viability and inflammatory responses in primary microglia. However, the fragment 51–81 exhibited impaired affinity to oligomeric Aβ. When administrated to the 5xFAD mice brain, the sTREM2 fragment 41–81, but not 51–81, increased the number of plaque-associated microglia and reduced the plaque deposition. Interestingly, the fragment 41–81 was more efficient than the physiological form of sTREM2 in ameliorating Aβ-related pathology. Conclusions Our results indicate that the interaction of sTREM2 truncated variants with Aβ is essential for enhancing microglial recruitment to the vicinity of an amyloid plaque and reducing the plaque load. Importantly, we identified a 41-amino acid sequence of sTREM2 that is sufficient for modulating microglial functions and more potent than the full-length sTREM2 in reducing the plaque load and the plaque-associated neurotoxicity. Taken together, our data provide more insights into the mechanisms underlying sTREM2 function and the minimal active sTREM2 sequence represents a promising candidate for AD therapy.
Collapse
Affiliation(s)
- Xuan Sheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yunling Yao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Ruizhi Huang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Ying Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yifei Zhu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Linting Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Lianshuai Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wanbing Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Rengong Zhuo
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, 361102, China.,Shenzhen Research Institute of Xiamen University, Shenzhen, 518063, China
| | - Dan Can
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Che-Feng Chang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China. .,Shenzhen Research Institute of Xiamen University, Shenzhen, 518063, China.
| |
Collapse
|
128
|
Hu B, Duan S, Wang Z, Li X, Zhou Y, Zhang X, Zhang YW, Xu H, Zheng H. Insights Into the Role of CSF1R in the Central Nervous System and Neurological Disorders. Front Aging Neurosci 2021; 13:789834. [PMID: 34867307 PMCID: PMC8634759 DOI: 10.3389/fnagi.2021.789834] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/26/2021] [Indexed: 01/15/2023] Open
Abstract
The colony-stimulating factor 1 receptor (CSF1R) is a key tyrosine kinase transmembrane receptor modulating microglial homeostasis, neurogenesis, and neuronal survival in the central nervous system (CNS). CSF1R, which can be proteolytically cleaved into a soluble ectodomain and an intracellular protein fragment, supports the survival of myeloid cells upon activation by two ligands, colony stimulating factor 1 and interleukin 34. CSF1R loss-of-function mutations are the major cause of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) and its dysfunction has also been implicated in other neurodegenerative disorders including Alzheimer’s disease (AD). Here, we review the physiological functions of CSF1R in the CNS and its pathological effects in neurological disorders including ALSP, AD, frontotemporal dementia and multiple sclerosis. Understanding the pathophysiology of CSF1R is critical for developing targeted therapies for related neurological diseases.
Collapse
Affiliation(s)
- Banglian Hu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Shengshun Duan
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Ziwei Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Xin Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Yuhang Zhou
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China
| | - Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Institute of Neuroscience, Xiamen University, Xiamen, China.,Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
129
|
Doroszkiewicz J, Mroczko P, Kulczyńska-Przybik A. Inflammation in the CNS - understanding various aspects of the pathogenesis of Alzheimer's disease. Curr Alzheimer Res 2021; 19:16-31. [PMID: 34856902 PMCID: PMC9127729 DOI: 10.2174/1567205018666211202143935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is a progressive and deadly neurodegenerative disorder, and one of the most common causes of dementia in the world. Current, insufficiently sensitive and specific methods of early diagnosis and monitoring of this disease prompt a search for new tools. Numerous literature data indicate that the pathogenesis of Alzheimer's disease (AD) is not limited to the neuronal compartment, but involves various immunological mechanisms. Neuroinflammation has been recognized as a very important process in AD pathology. It seems to play pleiotropic roles, both neuroprotective as well as neurodegenerative, in the development of cognitive impairment depending on the stage of the disease. Mounting evidence demonstrates that inflammatory proteins could be considered biomarkers of disease progression. Therefore, the present review summarizes the role of some inflammatory molecules and their potential utility in the detection and monitoring of dementia severity. The paper also provides a valuable insight into new mechanisms leading to the development of dementia, which might be useful in discovering possible anti-inflammatory treatment.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok. Poland
| | - Piotr Mroczko
- Department of Criminal Law and Criminology, Faculty of Law, University of Bialystok, Bialystok. Poland
| | | |
Collapse
|
130
|
Li Y, Laws SM, Miles LA, Wiley JS, Huang X, Masters CL, Gu BJ. Genomics of Alzheimer's disease implicates the innate and adaptive immune systems. Cell Mol Life Sci 2021; 78:7397-7426. [PMID: 34708251 PMCID: PMC11073066 DOI: 10.1007/s00018-021-03986-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/14/2021] [Accepted: 10/16/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterised by cognitive impairment, behavioural alteration, and functional decline. Over 130 AD-associated susceptibility loci have been identified by genome-wide association studies (GWAS), while whole genome sequencing (WGS) and whole exome sequencing (WES) studies have identified AD-associated rare variants. These variants are enriched in APOE, TREM2, CR1, CD33, CLU, BIN1, CD2AP, PILRA, SCIMP, PICALM, SORL1, SPI1, RIN3, and more genes. Given that aging is the single largest risk factor for late-onset AD (LOAD), the accumulation of somatic mutations in the brain and blood of AD patients have also been explored. Collectively, these genetic findings implicate the role of innate and adaptive immunity in LOAD pathogenesis and suggest that a systemic failure of cell-mediated amyloid-β (Aβ) clearance contributes to AD onset and progression. AD-associated variants are particularly enriched in myeloid-specific regulatory regions, implying that AD risk variants are likely to perturbate the expression of myeloid-specific AD-associated genes to interfere Aβ clearance. Defective phagocytosis, endocytosis, and autophagy may drive Aβ accumulation, which may be related to naturally-occurring antibodies to Aβ (Nabs-Aβ) produced by adaptive responses. Passive immunisation is providing efficiency in clearing Aβ and slowing cognitive decline, such as aducanumab, donanemab, and lecanemab (ban2401). Causation of AD by impairment of the innate immunity and treatment using the tools of adaptive immunity is emerging as a new paradigm for AD, but immunotherapy that boosts the innate immune functions of myeloid cells is highly expected to modulate disease progression at asymptomatic stage.
Collapse
Affiliation(s)
- Yihan Li
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Dr, Joondalup, WA, 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Dr, Joondalup, WA, 6027, Australia
| | - Luke A Miles
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - James S Wiley
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Xin Huang
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Ben J Gu
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
131
|
Bettcher BM, Tansey MG, Dorothée G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease - a research prospectus. Nat Rev Neurol 2021; 17:689-701. [PMID: 34522039 PMCID: PMC8439173 DOI: 10.1038/s41582-021-00549-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Dysregulation of the immune system is a cardinal feature of Alzheimer disease (AD), and a considerable body of evidence indicates pathological alterations in central and peripheral immune responses that change over time. Considering AD as a systemic immune process raises important questions about how communication between the peripheral and central compartments occurs and whether this crosstalk represents a therapeutic target. We established a whitepaper workgroup to delineate the current status of the field and to outline a research prospectus for advancing our understanding of peripheral-central immune crosstalk in AD. To guide the prospectus, we begin with an overview of seminal clinical observations that suggest a role for peripheral immune dysregulation and peripheral-central immune communication in AD, followed by formative animal data that provide insights into possible mechanisms for these clinical findings. We then present a roadmap that defines important next steps needed to overcome conceptual and methodological challenges, opportunities for future interdisciplinary research, and suggestions for translating promising mechanistic studies into therapeutic interventions.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Behavioral Neurology Section, Department of Neurology, University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Malú G Tansey
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
| | - Guillaume Dorothée
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Team "Immune System and Neuroinflammation", Hôpital Saint-Antoine, Paris, France
| | - Michael T Heneka
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
132
|
Rodriguez-Vieitez E, Ashton NJ. Plasma sTREM2: a potential marker of cerebrovascular injury in neurodegenerative disorders. Brain 2021; 144:3283-3285. [PMID: 34672320 PMCID: PMC8677507 DOI: 10.1093/brain/awab399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/16/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| |
Collapse
|
133
|
Discovery of Active Ingredients Targeted TREM2 by SPR Biosensor-UPLC/MS Recognition System, and Investigating the Mechanism of Anti-Neuroinflammatory Activity on the Lignin-Amides from Datura metel Seeds. Molecules 2021; 26:molecules26195946. [PMID: 34641490 PMCID: PMC8512677 DOI: 10.3390/molecules26195946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
As a new target protein for Alzheimer’s disease (AD), the triggering receptor expressed on myeloid Cells 2 (TREM2) was expressed on the surface of microglia, which was shown to regulate neuroinflammation, be associated with a variety of neuropathologic, and regarded as a potential indicator for monitoring AD. In this study, a novel recognition system based on surface plasmon resonance (SPR) for the TREM2 target spot was established coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC-MS), in order to screen the active ingredients targeting TREM2 from Datura metel seeds. The results showed that four lignan-amides were discovered as candidate compounds by SPR biosensor-UPLC/MS recognition analysis. According to the guidance of the active ingredients discovered by the system, the lignin-amides from Datura metel seeds (LDS) were preliminarily identified as containing 27 lignan-amides, which were enriched compositions by the HP-20 of Datura metel seeds. Meanwhile, the anti-inflammatory activity of LDS was evaluated in BV2 microglia induced by LPS. Our experimental results demonstrated that LDS could reduce NO release in LPS-treated BV2 microglia cells and significantly reduce the expression of the proteins of inducible Nitric Oxide Synthase (iNOS), cyclooxygenase 2 (COX-2), microtubule-associated protein tau (Tau), and ionized calcium-binding adapter molecule 1 (IBA-1). Accordingly, LDS might increase the expression of TREM2/DNAX-activating protein of 12 kDa (DAP12) and suppress the Toll-like receptor SX4 (TLR4) pathway and Recombinant NLR Family, Pyrin Domain Containing Protein 3 (NLRP3)/cysteinyl aspartate specific proteinase-1 (Caspase-1) inflammasome expression by LDS in LPS-induced BV2 microglial cells. Then, the inhibitory release of inflammatory factors Interleukin 1 beta (IL-1β), Interleukin 6 (IL-6), and Tumor necrosis factor-alpha (TNFα) inflammatory cytokines were detected to inhibit neuroinflammatory responses. The present results propose that LDS has potential as an anti-neuroinflammatory agent against microglia-mediated neuroinflammatory disorders.
Collapse
|
134
|
Tsai HH, Chen YF, Yen RF, Lo YL, Yang KC, Jeng JS, Tsai LK, Chang CF. Plasma soluble TREM2 is associated with white matter lesions independent of amyloid and tau. Brain 2021; 144:3371-3380. [PMID: 34515756 DOI: 10.1093/brain/awab332] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cerebral small vessel disease is one of the most common causes of cognitive decline and stroke. While several lines of evidence have established a relationship between inflammation and cerebrovascular pathology, the mechanistic link has not yet been elucidated. Recent studies suggest activation of immune mediators, including the soluble form of triggering receptor expressed on myeloid cells 2 (TREM2), may be critical regulators. In this study, we compared the plasma levels of soluble TREM2 and its correlations with neuroimaging markers and cerebral amyloid load in ten patients with Alzheimer's disease and 66 survivors of spontaneous intracerebral haemorrhage with cerebral amyloid angiopathy or hypertensive small vessel disease, two of the most common types of sporadic small vessel disease. We performed brain MRI and 11C-Pittsburgh compound B PET for all participants to evaluate radiological small vessel disease markers and cerebral amyloid burden, and 18F-T807 PET in a subgroup of patients to evaluate cortical tau pathology. Plasma soluble TREM2 levels were comparable between patients with Alzheimer's disease and small vessel disease (P=0.690). In patients with small vessel disease, plasma soluble TREM2 was significantly associated with white matter hyperintensity volume (P<0.001), but not with cerebral amyloid load. Among patients with Alzheimer's disease and cerebral amyloid angiopathy, plasma soluble TREM2 was independently associated with a tau-positive scan (P=0.001) and white matter hyperintensity volume (P=0.013), but not amyloid load (P=0.221). Our results indicate plasma soluble TREM2 is associated with white matter hyperintensity independent of amyloid and tau pathology. These findings highlight the potential utility of plasma soluble TREM2 as a strong predictive marker for small vessel disease-related white matter injury and hold clinical implications for targeting the innate immune response when treating this disease.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan.,Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Ling Lo
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
135
|
Wu KM, Zhang YR, Huang YY, Dong Q, Tan L, Yu JT. The role of the immune system in Alzheimer's disease. Ageing Res Rev 2021; 70:101409. [PMID: 34273589 DOI: 10.1016/j.arr.2021.101409] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder where the accumulation of amyloid plaques and the formation of tau tangles are the prominent pathological hallmarks. Increasing preclinical and clinical studies have revealed that different components of the immune system may act as important contributors to AD etiology and pathogenesis. The recognition of misfolded Aβ and tau by immune cells can trigger a series of complex immune responses in AD, and then lead to neuroinflammation and neurodegeneration. In parallel, genome-wide association studies have also identified several immune related loci associated with increased - risk of AD by interfering with the function of immune cells. Other immune related factors, such as impaired immunometabolism, defective meningeal lymphatic vessels and autoimmunity might also be involved in the pathogenesis of AD. Here, we review the data showing the alterations of immune cells in the AD trajectory and seek to demonstrate the crosstalk between the immune cell dysfunction and AD pathology. We then discuss the most relevant research findings in regards to the influences of gene susceptibility of immune cells for AD. We also consider impaired meningeal lymphatics, immunometabolism and autoimmune mechanisms in AD. In addition, immune related biomarkers and immunotherapies for AD are also mentioned in order to offer novel insights for future research.
Collapse
|
136
|
Choi H, Choi Y, Lee EJ, Kim H, Lee Y, Kwon S, Hwang DW, Lee DS. Hippocampal glucose uptake as a surrogate of metabolic change of microglia in Alzheimer's disease. J Neuroinflammation 2021; 18:190. [PMID: 34465358 PMCID: PMC8408933 DOI: 10.1186/s12974-021-02244-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/18/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Dynamically altered microglia play an important role in the progression of Alzheimer's disease (AD). Here, we found a close association of the metabolic reconfiguration of microglia with increased hippocampal glucose uptake on [18F]fluorodeoxyglucose (FDG) PET. METHODS We used an AD animal model, 5xFAD, to analyze hippocampal glucose metabolism using both animal FDG PET and ex vivo FDG uptake test. Cells of the hippocampus were isolated to perform single-cell RNA-sequencing (scRNA-seq). The molecular features of cells associated with glucose metabolism were analyzed at a single-cell level. In order to apply our findings to human brain imaging study, brain FDG PET data obtained from the Alzheimer's Disease Neuroimaging Initiative were analyzed. FDG uptake in the hippocampus was compared according to the diagnosis, AD, mild cognitive impairment, and controls. The correlation analysis between hippocampal FDG uptake and soluble TREM2 in cerebrospinal fluid was performed. RESULTS In the animal study, 8- and 12-month-old 5xFAD mice showed higher FDG uptake in the hippocampus than wild-type mice. Cellular FDG uptake tests showed that FDG activity in hippocampal microglia was increased in the AD model, while FDG activity in non-microglial cells of the hippocampus was not different between the AD model and wild-type. scRNA-seq data showed that changes in glucose metabolism signatures including glucose transporters, glycolysis and oxidative phosphorylation, mainly occurred in microglia. A subset of microglia with higher glucose transporters with defective glycolysis and oxidative phosphorylation was increased according to disease progression. In the human imaging study, we found a positive association between soluble TREM2 and hippocampal FDG uptake. FDG uptake in the hippocampus at the baseline scan predicted mild cognitive impairment conversion to AD. CONCLUSIONS We identified the reconfiguration of microglial glucose metabolism in the hippocampus of AD, which could be evaluated by FDG PET as a feasible surrogate imaging biomarker for microglia-mediated inflammation.
Collapse
Affiliation(s)
- Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, Jongo-Gu 03080 Republic of Korea
| | - Yoori Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Eun Ji Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Hyun Kim
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Youngsun Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Seokjun Kwon
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Do Won Hwang
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, Jongo-Gu 03080 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Department of Nuclear Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, Jongo-Gu 03080 Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 101 Daehak-ro, Jongno-Gu, Seoul 03080 Seoul, Republic of Korea
| |
Collapse
|
137
|
Ibanez L, Cruchaga C, Fernández MV. Advances in Genetic and Molecular Understanding of Alzheimer's Disease. Genes (Basel) 2021; 12:1247. [PMID: 34440421 PMCID: PMC8394321 DOI: 10.3390/genes12081247] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 01/19/2023] Open
Abstract
Alzheimer's disease (AD) has become a common disease of the elderly for which no cure currently exists. After over 30 years of intensive research, we have gained extensive knowledge of the genetic and molecular factors involved and their interplay in disease. These findings suggest that different subgroups of AD may exist. Not only are we starting to treat autosomal dominant cases differently from sporadic cases, but we could be observing different underlying pathological mechanisms related to the amyloid cascade hypothesis, immune dysfunction, and a tau-dependent pathology. Genetic, molecular, and, more recently, multi-omic evidence support each of these scenarios, which are highly interconnected but can also point to the different subgroups of AD. The identification of the pathologic triggers and order of events in the disease processes are key to the design of treatments and therapies. Prevention and treatment of AD cannot be attempted using a single approach; different therapeutic strategies at specific disease stages may be appropriate. For successful prevention and treatment, biomarker assays must be designed so that patients can be more accurately monitored at specific points during the course of the disease and potential treatment. In addition, to advance the development of therapeutic drugs, models that better mimic the complexity of the human brain are needed; there have been several advances in this arena. Here, we review significant, recent developments in genetics, omics, and molecular studies that have contributed to the understanding of this disease. We also discuss the implications that these contributions have on medicine.
Collapse
Affiliation(s)
- Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA; (L.I.); (C.C.)
- Neurogenomics and Informatics Center, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA; (L.I.); (C.C.)
- Neurogenomics and Informatics Center, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660 S. Euclid Ave. B8111, St. Louis, MO 63110, USA
| | - Maria Victoria Fernández
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA; (L.I.); (C.C.)
- Neurogenomics and Informatics Center, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO 63110, USA
| |
Collapse
|
138
|
Biel D, Brendel M, Rubinski A, Buerger K, Janowitz D, Dichgans M, Franzmeier N. Tau-PET and in vivo Braak-staging as prognostic markers of future cognitive decline in cognitively normal to demented individuals. ALZHEIMERS RESEARCH & THERAPY 2021; 13:137. [PMID: 34384484 PMCID: PMC8361801 DOI: 10.1186/s13195-021-00880-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022]
Abstract
Background To systematically examine the clinical utility of tau-PET and Braak-staging as prognostic markers of future cognitive decline in older adults with and without cognitive impairment. Methods In this longitudinal study, we included 396 cognitively normal to dementia subjects with 18F-Florbetapir/18F-Florbetaben-amyloid-PET, 18F-Flortaucipir-tau-PET and ~ 2-year cognitive follow-up. Annual change rates in global cognition (i.e., MMSE, ADAS13) and episodic memory were calculated via linear-mixed models. We determined global amyloid-PET (Centiloid) plus global and Braak-stage-specific tau-PET SUVRs, which were stratified as positive(+)/negative(−) at pre-established cut-offs, classifying subjects as Braak0/BraakI+/BraakI–IV+/BraakI–VI+/Braakatypical+. In bootstrapped linear regression, we assessed the predictive accuracy of global tau-PET SUVRs vs. Centiloid on subsequent cognitive decline. To test for independent tau vs. amyloid effects, analyses were further controlled for the contrary PET-tracer. Using ANCOVAs, we tested whether more advanced Braak-stage predicted accelerated future cognitive decline. All models were controlled for age, sex, education, diagnosis, and baseline cognition. Lastly, we determined Braak-stage-specific conversion risk to mild cognitive impairment (MCI) or dementia. Results Baseline global tau-PET SUVRs explained more variance (partial R2) in future cognitive decline than Centiloid across all cognitive tests (Cohen’s d ~ 2, all tests p < 0.001) and diagnostic groups. Associations between tau-PET and cognitive decline remained consistent when controlling for Centiloid, while associations between amyloid-PET and cognitive decline were non-significant when controlling for tau-PET. More advanced Braak-stage was associated with gradually worsening future cognitive decline, independent of Centiloid or diagnostic group (p < 0.001), and elevated conversion risk to MCI/dementia. Conclusion Tau-PET and Braak-staging are highly predictive markers of future cognitive decline and may be promising single-modality estimates for prognostication of patient-specific progression risk in clinical settings. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00880-x.
Collapse
Affiliation(s)
- Davina Biel
- Institute, for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, 80336, Munich, Germany
| | - Anna Rubinski
- Institute, for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Katharina Buerger
- Institute, for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Daniel Janowitz
- Institute, for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany
| | - Martin Dichgans
- Institute, for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute, for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377, Munich, Germany.
| | | |
Collapse
|
139
|
Moutinho M, Landreth GE. TREM2 splicing emerges as crucial aspect to understand TREM2 biology. J Leukoc Biol 2021; 110:827-828. [PMID: 34312904 DOI: 10.1002/jlb.2ce0521-264r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/05/2022] Open
Abstract
Discussion on TREM2 splicing and its different transcripts with regard to the physiological function of TREM2 and its role in disease.
Collapse
Affiliation(s)
- Miguel Moutinho
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Gary E Landreth
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
140
|
Qu Y, Ma YH, Huang YY, Ou YN, Shen XN, Chen SD, Dong Q, Tan L, Yu JT. Blood biomarkers for the diagnosis of amnestic mild cognitive impairment and Alzheimer's disease: A systematic review and meta-analysis. Neurosci Biobehav Rev 2021; 128:479-486. [PMID: 34245759 DOI: 10.1016/j.neubiorev.2021.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/06/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
The development of blood-based biomarkers of Alzheimer's disease (AD) pathology as tools for screening the general population is essential, but persists controversies. We aimed to evaluate the effects of AD core pathological biomarkers on blood, and systematical searched Embase, PubMed and Cochrane for eligible studies. Biomarker performance was rated by random-effects meta-analysis based on the ratio of means method and multivariable-adjusted effect estimates. Finally, 150 articles were included, which demonstrated T-tau (average ratio: 1.25-1.62), P-tau 181 (1.36-2.16) and NfL (1.24-1.86) were increased, and AβPPr (0.65-0.88) were decreased from controls to amnestic mild cognitive impairment (aMCI) to AD. Furthermore, Aβ42, Aβ42/Aβ40 ratio and P-tau 217 using ultrasensitive platforms also had great diagnostic accuracy from controls to aMCI to AD. Consequently, significantly changes of blood AD core biomarkers were verified in comparison between AD, aMCI and control, supporting biomarkers were strongly valid in identifying AD and aMCI, which provides a new prospect of AD early diagnosis and progressive monitoring. This study is registered with PROSPERO, number CRD42020191927.
Collapse
Affiliation(s)
- Yi Qu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Yuan Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
141
|
Zhang PF, Hu H, Tan L, Yu JT. Microglia Biomarkers in Alzheimer's Disease. Mol Neurobiol 2021; 58:3388-3404. [PMID: 33713018 DOI: 10.1007/s12035-021-02348-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Early detection and clinical diagnosis of Alzheimer's disease (AD) have become an extremely important link in the prevention and treatment of AD. Because of the occult onset, the diagnosis and treatment of AD based on clinical symptoms are increasingly challenged by current severe situations. Therefore, molecular diagnosis models based on early AD pathological markers have received more attention. Among the possible pathological mechanisms, microglia which are necessary for normal brain function are highly expected and have been continuously studied in various models. Several AD biomarkers already exist, but currently there is a paucity of specific and sensitive microglia biomarkers which can accurately measure preclinical AD. Bringing microglia biomarkers into the molecular diagnostic system which is based on fluid and neuroimaging will play an important role in future scientific research and clinical practice. Furthermore, developing novel, more specific, and sensitive microglia biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial microglial functions in response to AD pathology. This review discusses microglia biomarkers in the context of AD.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
142
|
Pillai JA, Khrestian M, Bena J, Leverenz JB, Bekris LM. Temporal Ordering of Inflammatory Analytes sTNFR2 and sTREM2 in Relation to Alzheimer's Disease Biomarkers and Clinical Outcomes. Front Aging Neurosci 2021; 13:676744. [PMID: 34276339 PMCID: PMC8279003 DOI: 10.3389/fnagi.2021.676744] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory changes are among the key markers of Alzheimer's disease (AD) related pathological changes. Pro-inflammatory analytes have been related to cognitive decline while others have been related to attenuating neuronal death. Among them, changes in cerebrospinal fluid (CSF) levels of soluble triggering receptor expressed on myeloid cells 2 (sTREM2) and soluble tumor necrosis factor receptor 2 (sTNFR2) have been described as impacting favorable clinical outcomes in AD. We therefore evaluate the effect of CSF sTREM2 and sTNFR2 when taken together on AD biomarkers and longitudinal clinical decline to understand their relative role on impacting AD clinical biomarkers and subsequent clinical outcomes. This longitudinal observational cohort study included 168 amyloid-positive (A+) and p-tau-positive (T+) participants with mild cognitive impairment (MCI) or AD dementia from the Alzheimer's Disease Neuroimaging Initiative (ADNI) with 109 of them having concomitant CSF sTREM2 and sTNFR2 data and 48 A+ T+ participants with MCI from a tertiary memory clinic cohort. An exploratory analysis was performed using data from 86 cognitively normal (CN) participants from ADNI with 72 of them having concomitant CSF AD biomarkers and CSF sTREM2 and sTNFR2 data. General linear models were used to evaluate the effect of sTREM2 and sTNFR2 levels on baseline CSF Aβ42, t-tau, and p-tau, and a linear mixed-effects model was used to assess longitudinal cognitive change after controlling for well-known covariates. Among ADNI A+ T+ MCI and AD dementia participants, CSF sTNFR2 had a stronger association, than CSF sTREM2, with CSF t-tau and p-tau. This was replicated among A+ T+ MCI participants from the memory clinic cohort. On the contrary, among A+ T+ CN participants, CSF sTREM2 explained significant variance in CSF t-tau and p-tau, while CSF sTNFR2 did not. When the effects of CSF sTNFR2 and t-tau on longitudinal cognitive change were taken into account, higher CSF sTREM2 predicted slower cognitive decline in A+ T+ AD dementia participants and faster decline in A+ T+ CN participants. Our results show that given the dynamic changes in sTREM2 and sTNFR2, the clinical impact of these distinct inflammation related biomarkers in tracking AD temporal progression across disease stages are likely to differ.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Department of Neurology, Cleveland Clinic, Cleveland, OH, United States
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Maria Khrestian
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - James B. Leverenz
- Department of Neurology, Cleveland Clinic, Cleveland, OH, United States
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
143
|
Hu WT, Ozturk T, Kollhoff A, Wharton W, Christina Howell J. Higher CSF sTNFR1-related proteins associate with better prognosis in very early Alzheimer's disease. Nat Commun 2021; 12:4001. [PMID: 34183654 PMCID: PMC8238986 DOI: 10.1038/s41467-021-24220-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is associated with Alzheimer's disease, but the application of cerebrospinal fluid measures of inflammatory proteins may be limited by overlapping pathways and relationships between them. In this work, we measure 15 cerebrospinal proteins related to microglial and T-cell functions, and show them to reproducibly form functionally-related groups within and across diagnostic categories in 382 participants from the Alzheimer's Disease Neuro-imaging Initiative as well participants from two independent cohorts. We further show higher levels of proteins related to soluble tumor necrosis factor receptor 1 are associated with reduced risk of conversion to dementia in the multi-centered (p = 0.027) and independent (p = 0.038) cohorts of people with mild cognitive impairment due to predicted Alzheimer's disease, while higher soluble TREM2 levels associated with slower decline in the dementia stage of Alzheimer's disease. These inflammatory proteins thus provide prognostic information independent of established Alzheimer's markers.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA.
- Rutgers Robert Wood Johnson Medical School and Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, USA.
| | - Tugba Ozturk
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | - Alexander Kollhoff
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | - Whitney Wharton
- Nell Hodgson School of Nursing, Emory University, Atlanta, GA, USA
| | - J Christina Howell
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
144
|
Yang HS, Zhang C, Carlyle BC, Zhen SY, Trombetta BA, Schultz AP, Pruzin JJ, Fitzpatrick CD, Yau WYW, Kirn DR, Rentz DM, Arnold SE, Johnson KA, Sperling RA, Chhatwal JP, Tanzi RE. Plasma IL-12/IFN-γ axis predicts cognitive trajectories in cognitively unimpaired older adults. Alzheimers Dement 2021; 18:645-653. [PMID: 34160128 DOI: 10.1002/alz.12399] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/02/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Immune dysregulation is implicated in neurodegeneration and altered cytokine levels are seen in people with dementia. However, whether cytokine levels are predictive of cognitive decline in cognitively unimpaired (CU) elderly, especially in the setting of elevated amyloid beta (Aβ), remains unclear. METHODS We measured nine cytokines in the baseline plasma of 298 longitudinally followed CU elderly and assessed whether these measures were associated with cognitive decline, alone or synergistically with Aβ. We next examined associations between cytokine levels and neuroimaging biomarkers of Aβ/tau/neurodegeneration. RESULTS Higher IL-12p70 was associated with slower cognitive decline in the setting of higher Aβ (false discovery rate [FDR] = 0.0023), whereas higher IFN-γ was associated with slower cognitive decline independent of Aβ (FDR = 0.013). Higher IL-12p70 was associated with less tau and neurodegeneration in participants with higher Aβ. DISCUSSION Immune dysregulation is implicated in early-stage cognitive decline, and greater IL-12/IFN-γ axis activation may be protective against cognitive decline and early-stage AD progression.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Can Zhang
- Harvard Medical School, Boston, Massachusetts, USA.,Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Becky C Carlyle
- Harvard Medical School, Boston, Massachusetts, USA.,Alzheimer's Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Sherri Y Zhen
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Bianca A Trombetta
- Alzheimer's Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jeremy J Pruzin
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Colleen D Fitzpatrick
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Wai-Ying W Yau
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Steven E Arnold
- Harvard Medical School, Boston, Massachusetts, USA.,Alzheimer's Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Rudolph E Tanzi
- Harvard Medical School, Boston, Massachusetts, USA.,Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| |
Collapse
|
145
|
The relationship of soluble TREM2 to other biomarkers of sporadic Alzheimer's disease. Sci Rep 2021; 11:13050. [PMID: 34158530 PMCID: PMC8219697 DOI: 10.1038/s41598-021-92101-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/28/2021] [Indexed: 11/16/2022] Open
Abstract
Microglial activation is a central player in the pathophysiology of Alzheimer’s disease (AD). The soluble fragment of triggering receptor expressed on myeloid cells 2 (sTREM2) can serve as a marker for microglial activation and has been shown to be overexpressed in AD. However, the relationship of sTREM2 with other AD biomarkers has not been extensively studied. We investigated the relationship between cerebrospinal fluid (CSF) sTREM2 and other AD biomarkers and examined the correlation of plasma sTREM2 with CSF sTREM2 in a cohort of individuals with AD and without AD. Participants were consecutively recruited from Asan Medical Center from 2018 to 2020. Subjects were stratified by their amyloid positivity and clinical status. Along with other AD biomarkers, sTREM2 level was measured in the plasma as well as CSF. In 101 patients with either amyloid-positive or negative status, CSF sTREM2 was closely associated with CSF T-tau and P-tau and not with Abeta42. CSF sTREM2 levels were found to be strongly correlated with CSF neurofilament light chain. The comparison of CSF and plasma sTREM2 levels tended to have an inverse correlation. Plasma sTREM2 and P-tau levels were oppositely influenced by age. Our results suggest that neuroinflammation may be closely associated with tau-induced neurodegeneration.
Collapse
|
146
|
Süß P, Schlachetzki JCM. Microglia in Alzheimer's Disease. Curr Alzheimer Res 2021; 17:29-43. [PMID: 32048973 DOI: 10.2174/1567205017666200212155234] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 12/31/2019] [Accepted: 01/20/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's Disease (AD) is the most frequent neurodegenerative disorder. Although proteinaceous aggregates of extracellular Amyloid-β (Aβ) and intracellular hyperphosphorylated microtubule- associated tau have long been identified as characteristic neuropathological hallmarks of AD, a disease- modifying therapy against these targets has not been successful. An emerging concept is that microglia, the innate immune cells of the brain, are major players in AD pathogenesis. Microglia are longlived tissue-resident professional phagocytes that survey and rapidly respond to changes in their microenvironment. Subpopulations of microglia cluster around Aβ plaques and adopt a transcriptomic signature specifically linked to neurodegeneration. A plethora of molecules and pathways associated with microglia function and dysfunction has been identified as important players in mediating neurodegeneration. However, whether microglia exert either beneficial or detrimental effects in AD pathology may depend on the disease stage. In this review, we summarize the current knowledge about the stage-dependent role of microglia in AD, including recent insights from genetic and gene expression profiling studies as well as novel imaging techniques focusing on microglia in human AD pathology and AD mouse models.
Collapse
Affiliation(s)
- Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universitat, Erlangen- Nürnberg, Germany
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0651, United States
| |
Collapse
|
147
|
Wang SY, Gong PY, E Y, Zhang YD, Jiang T. The Role of TREML2 in Alzheimer's Disease. J Alzheimers Dis 2021; 76:799-806. [PMID: 32568208 DOI: 10.3233/jad-200406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Late-onset Alzheimer's disease (AD) accounts for most of all AD casesand is currently considered a complex disorder caused by a combination of environmental and genetic factors. As an important family member of triggering receptor expressed on myeloid cells (TREM), TREM-like transcript 2 gene (TREML2) locates on human chromosome 6p21.1, a newly-identified hot zone for AD susceptibility, and encodes atransmembrane immune receptor. Emerging evidence implied a potential role of TREML2 in the susceptibility and pathogenesis of AD. Here, we review the recent literature about the association of TREML2 variants with AD risk and disease endophenotypes. Moreover, we summarize the latest findings regarding cellular localization and biological functions of TREML2 and speculate its possible role in AD pathogenesis. In addition, we discuss future research directions of TREML2 and AD.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Yuhua Hospital, Nanjing, China
| | - Peng-Yu Gong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Yuhua Hospital, Nanjing, China
| | - Yan E
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Yuhua Hospital, Nanjing, China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Yuhua Hospital, Nanjing, China.,School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Neurology, Nanjing Yuhua Hospital, Nanjing, China
| |
Collapse
|
148
|
Matejuk A, Vandenbark AA, Offner H. Cross-Talk of the CNS With Immune Cells and Functions in Health and Disease. Front Neurol 2021; 12:672455. [PMID: 34135852 PMCID: PMC8200536 DOI: 10.3389/fneur.2021.672455] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
The immune system's role is much more than merely recognizing self vs. non-self and involves maintaining homeostasis and integrity of the organism starting from early development to ensure proper organ function later in life. Unlike other systems, the central nervous system (CNS) is separated from the peripheral immune machinery that, for decades, has been envisioned almost entirely as detrimental to the nervous system. New research changes this view and shows that blood-borne immune cells (both adaptive and innate) can provide homeostatic support to the CNS via neuroimmune communication. Neurodegeneration is mostly viewed through the lens of the resident brain immune populations with little attention to peripheral circulation. For example, cognition declines with impairment of peripheral adaptive immunity but not with the removal of microglia. Therapeutic failures of agents targeting the neuroinflammation framework (inhibiting immune response), especially in neurodegenerative disorders, call for a reconsideration of immune response contributions. It is crucial to understand cross-talk between the CNS and the immune system in health and disease to decipher neurodestructive and neuroprotective immune mechanisms for more efficient therapeutic strategies.
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
| | - Arthur A Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, United States.,Department of Neurology, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, United States.,Department of Neurology, Oregon Health and Science University, Portland, OR, United States.,Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
149
|
Morgan DG, Mielke MM. Knowledge gaps in Alzheimer's disease immune biomarker research. Alzheimers Dement 2021; 17:2030-2042. [PMID: 33984178 DOI: 10.1002/alz.12342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 11/09/2022]
Abstract
Considerable evidence has accumulated implicating a role for immune mechanisms in moderating the pathology in Alzheimer's disease dementia. However, the appropriate therapeutic target, the appropriate direction of manipulation, and the stage of disease at which to begin treatment remain unanswered questions. Part of the challenge derives from the absence of any selective pressure to develop a coordinated beneficial immune response to severe neural injury in adults. Thus, immune responses to the prevailing stimuli are likely to contain both beneficial and detrimental components. Knowledge gaps include: (1) how a biomarker change relates to the underlying biology, (2) the degree to which pathological stage group differences reflect a response to pathology versus trait differences among individuals regulating risk of developing pathology, (3) the degree to which biomarker levels are predictive of subsequent changes in pathology and/or cognition, and (4) experimental manipulations in model systems to determine whether differences in immune biomarkers are causally related to pathology.
Collapse
Affiliation(s)
- David G Morgan
- Alzheimer's Alliance, Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
150
|
Rauchmann BS, Sadlon A, Perneczky R. Soluble TREM2 and Inflammatory Proteins in Alzheimer's Disease Cerebrospinal Fluid. J Alzheimers Dis 2021; 73:1615-1626. [PMID: 31958095 DOI: 10.3233/jad-191120] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The present study explores the associations of soluble TREM2, an important regulator of microglial activity linked to Alzheimer's disease (AD), with other known inflammatory proteins in cerebrospinal fluid (CSF). We studied 303 participants, including 89 controls, 135 mild cognitive impairment, and 79 AD dementia patients. Using established CSF biomarkers, subjects were classified according to the National Institute on Aging-Alzheimer's Association research framework, which groups markers into those of amyloid-β deposition (A), tau pathology (T), and neurodegeneration (N). TNFR1, TNFR2, TGF-β1, TGFβ2, IL-9, TNF-α, ICAM1, and VCAM1 showed significant concentration differences between the ATN groups, with higher concentrations in more advanced disease categories. sTREM2 was positively associated with the pro-inflammatory proteins TNF-α, TNFR1, TNFR2, ICAM1, VCAM1, and IP-10 and negatively with IL-21; also, positive associations with the anti-inflammatory proteins TGFβ1, IL-10, and IL-9 were found. Pathway enrichment analysis highlighted the involvement of sTREM2 in key functional clusters including immunoglobulin and cytokine production and cellular response to lipopolysaccharides, cytokines, and steroid hormones. Our work provides further evidence in support of TREM2 as amarker of neuroinflammatory response in AD.
Collapse
Affiliation(s)
- Boris-Stephan Rauchmann
- Department of Radiology, University Hospital, LMU Munich, Germany.,Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Germany
| | - Angélique Sadlon
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, UK
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Germany.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, UK.,German Center for Neurodegenerative Diseases (DZNE) Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | |
Collapse
|